CD90 exhibits cell type- and context-dependent expression:
Stem/Progenitor Cells: Mesenchymal stem cells (MSCs), hematopoietic stem cells (HSCs), keratinocytic stem cells (KSCs) .
Non-Lymphoid Cells: Fibroblasts, neurons, activated endothelial cells, and adventitial mesenchymal progenitor cells in blood vessels .
Immune Cells: Subsets of CD4+/CD8+ T cells (Th17/Tc17) in humans .
Cancer: Overexpressed in glioblastoma (GBM), hepatocellular carcinoma (HCC), melanoma, and breast cancer stem-like cells .
Vascular Disease: CD90+ adventitial mesenchymal progenitor cells show reduced angiogenic capacity in diseased vs. healthy human aortas .
Cell Adhesion/Migration: Facilitates leukocyte-endothelial interactions during inflammation and melanoma metastasis via αv/β3 integrin binding .
Neuronal Regulation: Inhibits neurite outgrowth in mature astrocytes .
Immune Modulation:
Stem Cell Isolation: CD90 enriches myogenic progenitor subpopulations in human urethral-derived cells (UDCs) .
Cardiac Regeneration:
Angiogenesis Modulation: CD90+ adventitial cells from healthy aortas improved limb perfusion in ischemic mice (perfusion ratio: 0.81 vs. 0.67 in diseased aorta cells) .
Cancer Therapy: CD90 knockdown reduces migration/invasion in GBM and HCC .
CD90 (also known as Thy-1) is a cell surface glycoprotein initially identified on mouse thymocytes. In humans, CD90 has been identified on various stem cells and at different expression levels in non-lymphoid tissues including fibroblasts, brain cells, and activated endothelial cells . Research has established specific expression patterns across human primary cells:
High expression: Normal Human Dermal Fibroblasts (NHDF), activated Human Umbilical Vein Endothelial Cells (HUVEC)
Low/No expression: Human Corneal Epithelial Cells (CEC), Retinal Pigmented Epithelial (RPE) cells, Normal Human Epidermal Keratinocytes (NHEK), Normal Human Bronchial Epithelial (NHBE) cells, non-activated HUVEC
This differential expression pattern makes CD90 a valuable biomarker for identifying specific cell populations in research and clinical applications.
The gold standard methodology for CD90 detection and quantification is flow cytometry using fluorescent-labeled anti-human CD90 monoclonal antibodies (α-CD90 MAb). The standard protocol includes:
Sample preparation: Trypsinization of adherent cells followed by washing with growth medium
Cell suspension preparation: Centrifugation at 200 × g for 5 min and resuspension in PBS with 2% FBS
Antibody staining: Incubation with fluorescein isothiocyanate-conjugated anti-human CD90 antibody (typically 1:200 dilution) for 30 min on ice in dark conditions
Viability staining: Addition of 7-AAD (1:100) for 10 min before analysis to exclude dead cells
Analysis: Final centrifugation, resuspension, filtration through a 70 μm filter, and analysis using flow cytometry
Additional methods include direct immunofluorescence for qualitative assessment and immunohistochemistry for tissue samples .
CD34+/CD90+ cells isolated from the stromal vascular fraction (SVF) of human adipose tissue exhibit multiple characteristic stem cell properties:
Sphere cluster formation: When grown in non-adherent conditions, these cells form sphere clusters, a hallmark of stem-like behavior
High proliferative capacity: They demonstrate superior proliferation compared to differentiated cells
Elevated telomerase activity: Their telomerase activity is significantly higher than in differentiated cells, indicating self-renewal potential
Side population phenotype: A fraction of these cells display the side population phenotype, associated with stemness
Multipotent differentiation: These cells can rapidly differentiate into adipocytes when cultured in adipogenic medium and into endothelial cells (CD31+/VEGF+/Flk-1+) when appropriately stimulated
Angiogenic potential: When placed in methylcellulose, they form capillary-like structures and produce high levels of VEGF, measurable by ELISA
These properties collectively establish CD34+/CD90+ adipose-derived stem cells as promising candidates for tissue reconstruction in regenerative medicine, particularly for vascular disease treatments .
Fibroblast contamination represents a significant challenge in maintaining specialized human primary cell cultures due to fibroblasts' rapid proliferation. CD90's differential expression can be utilized to develop an effective purification strategy:
Identification: First confirm fibroblast contamination through CD90 expression analysis using flow cytometry
Magnetic bead separation:
This method preserves the viability and functionality of the target cells while efficiently removing contaminating fibroblasts. This approach is particularly valuable for developing pure cell populations for cell therapy applications where maintaining specifically differentiated cultures is essential .
Meta-analysis data reveals significant correlations between CD90 overexpression and adverse clinical outcomes in cancer patients:
Sensitivity analysis confirmed that these associations remain statistically significant even after accounting for the influence of individual studies, establishing CD90 overexpression as a reliable prognostic biomarker for cancer progression and patient outcomes .
CD90 expression distinguishes functionally distinct fibro-adipogenic progenitor (FAP) subpopulations in human skeletal muscle, with significant implications for metabolic disorders:
CD90+ FAPs are larger in size
CD90+ FAPs proliferate faster
CD90+ FAPs express higher levels of extracellular matrix genes compared to CD90- FAPs
CD90+ FAPs are associated with muscle degeneration in Type 2 diabetic (T2D) patients
The dynamic relationship between CD90+ FAPs and insulin resistance appears significant, though the precise mechanisms require further investigation
CD90+ FAPs may contribute to the degenerative muscle microenvironment in metabolic disorders
Single-cell RNA sequencing analysis could further elucidate the heterogeneity of FAPs across healthy subjects, obese individuals, and T2D patients (both insulin-treated and non-treated). This approach would help clarify the molecular changes in FAP subpopulations during disease progression and potential therapeutic interventions .
CD90 has been identified as a crucial molecular signature for cells with myogenic differentiation potential. The following experimental workflow can assess this potential:
Isolation of CD90+ cells:
Myogenic induction:
Assessment of myogenic differentiation:
Research indicates that while CD90 itself doesn't promote differentiation, CD90+ cells respond more effectively to myogenic induction factors like MYOD1, suggesting the presence of additional factors in CD90+ cells that facilitate myogenic differentiation .
Single-cell RNA sequencing (scRNA-seq) offers powerful capabilities for investigating CD90+ cell populations:
Heterogeneity characterization:
Trajectory analysis:
Comparative analysis:
Regulatory network identification:
This comprehensive approach provides insights that traditional bulk analysis methods cannot deliver, particularly in understanding the complex heterogeneity and dynamic nature of CD90+ cell populations in human tissues .
CD90-expressing cells offer significant potential for regenerative medicine applications:
Vascular regeneration:
CD34+/CD90+ adipose-derived stem cells (ASCs) demonstrate robust angiogenic potential
These cells form capillary-like structures in methylcellulose
They produce high levels of VEGF, promoting vascularization
Their endothelial differentiation capacity makes them suitable for vascular tissue engineering
Skeletal muscle regeneration:
Translational considerations:
These characteristics position CD90-expressing cells as promising candidates for tissue reconstruction strategies, particularly for patients requiring treatments for vascular disease and muscle disorders .
Several methodological challenges must be addressed when isolating and characterizing CD90+ cells for clinical applications:
Heterogeneity management:
Purification efficiency:
Functional assessment:
Clinical-grade processing:
Regulatory considerations:
Addressing these challenges through rigorous methodological development and standardization will facilitate the translation of CD90+ cell-based therapies from bench to bedside .
Despite significant advances in CD90 research, several critical questions remain unresolved:
Regulatory mechanisms:
Developmental origins:
Functional mechanisms:
Disease associations:
Therapeutic potential:
Addressing these questions will require integration of advanced technologies including single-cell transcriptomics, lineage tracing, and functional genomics approaches .
Emerging technologies offer transformative potential for advancing CD90 research:
Single-cell multi-omics:
Spatial transcriptomics:
CRISPR-based functional genomics:
Organoid and microphysiological systems:
Advanced computational modeling:
These technological advances will facilitate deeper understanding of CD90 biology and accelerate translation of findings into clinical applications for regenerative medicine and disease treatment .
Thy-1 was first discovered in 1964 by Reif and Allen during their search for heterologous antisera against mouse leukemia cells. It was initially identified as a thymocyte antigen and was named theta (θ) antigen. Later, it was renamed Thy-1 (THYmocyte differentiation antigen 1) due to its identification in thymocytes .
Thy-1 is involved in several critical biological functions:
Recent studies have shown that Thy-1 plays a crucial role in the initial stage of human cytomegalovirus (HCMV) infection. It mediates HCMV infection at the entry step and is important for infection that occurs at a low multiplicity of infection (m.o.i.). Thy-1 interacts with HCMV glycoproteins gB and gH, forming a complex that is important for virus entry .
Recombinant Thy-1 is produced using recombinant DNA technology, which allows for the expression of the protein in various host systems. This recombinant form is used in research to study the protein’s function and its role in various biological processes.