Abstract

Cultured human bone marrow stromal (mesenchymal) stem cells (hBM‐MSCs) are heterogenous cell populations exhibiting variable biological properties. Quantitative high‐content imaging technology allows identification of morphological markers at a single cell resolution that are determinant for cellular functions. We determined the morphological characteristics of cultured primary hBM‐MSCs and examined their predictive value for hBM‐MSC functionality. BM‐MSCs were isolated from 56 donors and characterized for their proliferative and differentiation potential. We correlated these data with cellular and nuclear morphological features determined by Operetta; a high‐content imaging system. Cell area, cell geometry, and nucleus geometry of cultured hBM‐MSCs exhibited significant correlation with expression of hBM‐MSC membrane markers: ALP, CD146, and CD271. Proliferation capacity correlated negatively with cell and nucleus area and positively with cytoskeleton texture features. In addition, in vitro differentiation to osteoblasts as well as in vivo heterotopic bone formation was associated with decreased ratio of nucleus width to length. Multivariable analysis applying a stability selection procedure identified nuclear geometry and texture as predictors for hBM‐MSCs differentiation potential to osteoblasts or adipocytes. Our data demonstrate that by employing a limited number of cell morphological characteristics, it is possible to predict the functional phenotype of cultured hBM‐MSCs and thus can be used as a screening test for “quality” of hBM‐MSCs prior their use in clinical protocols.

Highlights

  • Human bone marrow-derived stromal stem cells are non-hematopoietic, self-renewing, plastic adherent cells with the ability to differentiate into several mesodermal lineages including osteoblasts (OB) and adipocytes (AD).[1]

  • MSCs-like cells have been isolated from different tissues such as adipose tissues, placenta, and Wharton's jelly of umbilical cord, bone marrow is considered the standard source for MSCs with in vitro and in vivo bone-forming ability[1,5,6] and is the basis for their clinical use for enhancing bone tissue formation

  • Cells cultured on high micropillars exhibited altered nucleus shape that favored osteoblastic differentiation, whereas cells grown on smooth or on low micropillars exhibited less deformed nuclei which were associated with adipocyte differentiation.[24]

Read more

Summary

| INTRODUCTION

Human bone marrow-derived stromal (mesenchymal) stem cells (hBM-MSCs) are non-hematopoietic, self-renewing, plastic adherent cells with the ability to differentiate into several mesodermal lineages including osteoblasts (OB) and adipocytes (AD).[1]. The actin cytoskeleton plays a role in hBM-MSC lineage commitment,[25,26] and microtubules are key players in cell proliferation[27] and have been reported to contribute to osteoblast differentiation.[28] Alterations in cell shape affects actin stress fibers, which through intracellular signaling pathways initiate cell lineage commitment.[23,29] nucleus shape can be modulated by cytoskeletal fibers in response to extracellular forces that affect cellular differentiation.[30] Liu et al showed that culturing cells on surface with different topography could affect nuclear morphology and cell differentiation in rat BM-MSCs. Cells cultured on high micropillars exhibited altered nucleus shape that favored osteoblastic differentiation, whereas cells grown on smooth or on low micropillars exhibited less deformed nuclei which were associated with adipocyte differentiation.[24]

Significance statement
| MATERIALS AND METHODS
| RESULTS
| DISCUSSION
Findings
CONFLICT OF INTEREST
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call