scholarly journals Superiority of Adipose-derived CD34+ Cells over Adipose-derived Stem Cells in Promoting Ischamic Tissue Survival

Author(s):  
Yan-Jun Liu ◽  
Tian-Yu Zhang ◽  
Poh-Ching Tan ◽  
Yun Xie ◽  
Pei-Qi Zhang ◽  
...  

Abstract Background: Tissue ischemia usually leads to necrosis and is a threatening condition associated with reconstructive surgery. Promoting the survival of ischemic tissue is critical for improving clinical outcomes. Although various solutions based on stem cells have been reported, there are still limitations to clinical translation. The aim of this study was to develop an effective method to promote the survival of ischemic tissue. Methods: Adipose-derived CD34+ and CD34- cells were obtained by magnetic bead sorting from the stromal vascular faction (SVF). Adipose-derived stem cell (ADSC) were collected by subculture. The angiogenic capacities of CD34+ cells, CD34- cells and ADSC were evaluated in vitro by comparing mRNA and protein expression. Random axial flaps in nude mice were used to evaluate the efficacy of these cells in protecting tissue from necrosis. The effect of these cells in preventing inflammation was also evaluated. Results: Our data suggest that CD34+ cells expressed higher levels of angiogenetic factors and lower levels of inflammatory factors than the other cell types. More vessel branches were formed when human umbilical vein endothelial cells (HUVECs) were treated with conditioned medium from CD34+ cells than conditioned medium from the other cell types. Compared to ADSC, CD34+ cells showed significantly higher efficacy in promoting tissue survival. More CD31+ cells and higher levels of angiogenic factors were observed in tissues from the CD34+ Group than from the other Groups. Lower levels of the proinflammatory factors TNF-α and IL-1b and higher levels of anti-inflammatory factors were found in the CD34+ Group than in the other Groups.Conclusion: Adipose-derived CD34+ cells showed better efficacy in improving ischemic tissue survival than ADSC by reducing tissue inflammation and promoting angiogenesis. CD34+ cells can be obtained easily and may be suitable for clinical applications.

2021 ◽  
Author(s):  
Yan-Jun Liu ◽  
Tian-Yu Zhang ◽  
Poh-Ching Tan ◽  
Pei-Qi Zhang ◽  
Yun Xie ◽  
...  

Abstract Background: Tissue ischemia usually leads to necrosis and is a threatening condition associated with reconstructive surgery. Promoting the survival of ischemic tissue is critical for improving clinical outcomes. Although various solutions based on stem cells have been reported, there are still limitations to clinical translation. The aim of this study was to develop an effective method to promote the survival of ischemic tissue.Methods: Adipose-derived CD34+ and CD34- cells were obtained by magnetic bead sorting from the stromal vascular faction (SVF). Adipose-derived stem cells (ADSCs) were collected by subculture. The angiogenic capacities of CD34+ cells, CD34- cells and ADSCs were evaluated in vitro by comparing mRNA and protein expression. Random axial flaps in nude mice were used to evaluate the efficacy of these cells in protecting tissue from necrosis. The effect of these cells in preventing inflammation was also evaluated.Results: Our data suggest that CD34+ cells expressed higher levels of angiogenetic factors and lower levels of inflammatory factors than the other cell types. More vessel branches were formed when human umbilical vein endothelial cells (HUVECs) were treated with conditioned medium from CD34+ cells than conditioned medium from the other cell types. Compared to ADSCs, CD34+ cells showed significantly higher efficacy in promoting tissue survival. More CD31+ cells and higher levels of angiogenic factors were observed in tissues from the CD34+ group than in those from the other groups. Lower levels of the proinflammatory factors TNF-α and IL-1b and higher levels of anti-inflammatory factors were found in the CD34+ group than in the other groups.Conclusion: Adipose-derived CD34+ cells showed better efficacy in improving ischemic tissue survival than ADSCs by reducing tissue inflammation and promoting angiogenesis. CD34+ cells can be obtained easily and may be suitable for clinical applications.


Author(s):  
А.П. Лыков ◽  
А.М. Чернявский ◽  
О.В. Повещенко ◽  
А.В. Фомичев ◽  
М.А. Суровцева ◽  
...  

Аутологичные костномозговые стволовые клетки являются альтернативным способом терапии пациентов с сердечной недостаточностью. Цель работы - изучение фенотипа и функциональных свойств костномозговых мононуклеарных клеток (КМ-МНК) больных хронической сердечной недостаточностью (ХСН) до и после кратковременной экспозиции с эритропоэтином in vitro . Методика. КМ-МНК выделяли на градиенте плотности фиколл/верографин (r = 1,077 г/л). Фенотип КМ-МНК, клеточный цикл и апоптоз CD34+ клеток определяли до и после экспозиции с эритропоэтином на проточном цитометре. Пролиферативный потенциал КМ-МНК до и после экспозиции с эритропоэтином оценивали в спонтанном и стимулирующем тесте. Пролиферацию, миграцию и ангиогенный потенциал клеток EA.hy 929 изучали в тесте «раневого дефекта» монослоя клеток и на матригеле под влиянием 30% кондиционных сред от КМ-МНК. Результаты. Показано, что КМ-МНК представляют собой смесь гемопоэтических стволовых клеток (ГСК), эндотелиальных прогениторных клеток (ЭПК) на разных этапах созревания и дифференцировки, и мезенхимных стволовых клеток (МСК). Под действием эритропоэтина увеличивается количество CD34+ клеток в G0G1 фазе клеточного цикла, CD45+/EpoR+, CD31-/CD184+, CD31+/CD184+ и CD34+/CD184-, и уменьшается количество CD34+/CD133+, CD34+/EpoR-. Кондиционная среда от КМ-МНК способствует пролиферации, миграции и формированию сосудисто-подобных структур клетками EA.hy 929. Заключение. Полученные результаты свидетельствуют, что кратковременная экспозиция КМ-МНК больных ХСН задерживает CD34+ клетки в стадии покоя, увеличивает пул ЭПК, экспрессирующих «хоуминг» рецептор, а кондиционная среда от КМ-МНК стимулирует пролиферацию, миграцию и ангиогенный потенциал EA.hy 929, что следует учитывать при выборе методов усиления «приживаемости» клеточного трансплантата. Autologous bone marrow stem cells are an alternative therapy for patients with heart failure. The aim of this work was to study the phenotype and functional properties of bone marrow mononuclear cells (BM-MNCs) from patients with chronic heart failure (CHF) before and after a short-term in vitro exposure to erythropoietin. Methods. BM-MNCs were isolated using density gradient. The BM-MNC phenotype, cell cycle, and apoptosis of CD34+ cells were evaluated before and after exposure to erythropoietin with a flow cytometer. Proliferation of BM-MNCs before and after the erythropoietin exposure was evaluated in a spontaneous and stimulating test. Proliferation, migration, and angiogenic potential of EA.hy 929 cells were studied in the wound closure test and in the tubule formation test under the influence of 30% conditioned medium from BM-MNCs. Results. BM-MNCs represented a mixture of hematopoietic stem cells (HSCs), endothelial progenitor cells (EPCs) at different stages of maturation and differentiation, and mesenchymal stem cells (MSCs). Erythropoietin increased the number of CD34+ cells in the G0/G1 cell cycle phase, CD45+/EpoR+, CD31-/CD184+, CD31+/CD184+, and CD34+/CD184-, and decreased the number of CD34+/CD133+ and CD34+/EpoR. The erythropoietin exposure of BM-MNCs reduced their proliferative capacity. The BM-MNCs-conditioned medium promoted EA.hy 929 cell proliferation, migration, and formation of vascular-like structures. Conclusion. A short-term exposure of BM-MNCs delayed the resting stage of CD34+ cells, increased the pool of EPCs expressing the homing receptor while the BM-MNC conditioned medium stimulated EA.hy 929 proliferation, migration and tubule formation, which should be taken into account when selecting methods to enhance survival of cellular grafts.


2011 ◽  
Vol 23 (1) ◽  
pp. 253
Author(s):  
M. Bionaz ◽  
T. Jensen ◽  
E. Monaco ◽  
Z. Dymon ◽  
A. J. Maki ◽  
...  

We have previously shown that heterologous transplantation of porcine adipose-derived stem cells (ADSC) enhances bone healing. Freshly harvested ADSC are a heterogeneous population that contains several types of cells other than stem cells. The isolation of highly purified ADSC could be of clinical importance. In this study, we compared the in vitro growth characteristics and in vivo healing potential of ADSC unsorted or separated using CD34 as a marker. The ADSC were extracted from the back fat of 4 male pigs at 6 months of age. For the in vitro experiment, aliquots of the ADSC were sorted by magnetic beads (Miltenyi Biotec, Gladbach, Germany) into CD34-positive (CD34+) and CD34-negative (CD34–) cell populations. The unsorted ADSC (uADSC), plus the CD34+, CD34–, and a 50:50 mixture of CD34+ and CD34– (MIX) were plated in 24-well plates and differentiated into osteocytes. A robotic stage inverted microscope was used to photograph the entire culture well, and then number, dimension, and density of bone nodules were assessed. Alizarin red (AR) staining was performed and quantified. Cells were harvested before cell plating and then on several time points during expansion, at confluence, and at 3, 6, or 18 days [d] of differentiation for cell counting and RNA extraction. Real-time RT-PCR was performed for CD34, COL1A1, and SPARC genes. For the in vivo experiment, freshly isolated ADSC were sorted by flow cytometry into CD34+ and CD34– cell populations. Unsorted and sorted cells were transplanted, in duplicate, into 10- or 25-mm mandible osteoctomies. Mandibles were harvested after 8 weeks for evaluation of healing by DEXA scanning. In vitro data were statistically analysed using a mixed model (SAS) with time and cell type as fixed effect and pig as the random effect. The in vivo data were assessed by ANOVA with cell types as the fixed effect and pig as the random effect. Freshly harvested ADSC contained 42.3 ± 11.0% CD34+ cells. The uADSC reached confluence at 6 days after plating, whereas other cell types reached confluence at 16 days. Expression of CD34 decreased after plating but was similar between cell types. Among osteogenic genes, only expression of SPARC increased during differentiation. The number of osteogenic nodules was higher (P < 0.05) in uADSC than the in other cell types, but the area and nodule density were similar to CD34– and greater (P < 0.05) than CD34+ and MIX. The amount of AR was higher (P < 0.05) in uADSC compared with CD34– and MIX but similar to CD34+. In the in vivo trial, uADSC had a greater (P < 0.05) healing compared with sorted cells. Among those, CD34– cells appeared to increase healing compared with CD34+ cells. Results indicate that CD34+ cells do not differ significantly from CD34– in the in vitro osteogenic differentiation but have lower in vivo healing capacity; however, in vitro data were confused by a lack of pure CD34– cells. The freshly isolated ADSC have a greater healing capacity than sorted cells, as indicated by in vitro and in vivo experiments. Overall our data indicate that the sorting of ADSC CD34+ cells is not of clinical relevance.


2016 ◽  
Vol 113 (8) ◽  
pp. 2206-2211 ◽  
Author(s):  
Xuanyi Ma ◽  
Xin Qu ◽  
Wei Zhu ◽  
Yi-Shuan Li ◽  
Suli Yuan ◽  
...  

The functional maturation and preservation of hepatic cells derived from human induced pluripotent stem cells (hiPSCs) are essential to personalized in vitro drug screening and disease study. Major liver functions are tightly linked to the 3D assembly of hepatocytes, with the supporting cell types from both endodermal and mesodermal origins in a hexagonal lobule unit. Although there are many reports on functional 2D cell differentiation, few studies have demonstrated the in vitro maturation of hiPSC-derived hepatic progenitor cells (hiPSC-HPCs) in a 3D environment that depicts the physiologically relevant cell combination and microarchitecture. The application of rapid, digital 3D bioprinting to tissue engineering has allowed 3D patterning of multiple cell types in a predefined biomimetic manner. Here we present a 3D hydrogel-based triculture model that embeds hiPSC-HPCs with human umbilical vein endothelial cells and adipose-derived stem cells in a microscale hexagonal architecture. In comparison with 2D monolayer culture and a 3D HPC-only model, our 3D triculture model shows both phenotypic and functional enhancements in the hiPSC-HPCs over weeks of in vitro culture. Specifically, we find improved morphological organization, higher liver-specific gene expression levels, increased metabolic product secretion, and enhanced cytochrome P450 induction. The application of bioprinting technology in tissue engineering enables the development of a 3D biomimetic liver model that recapitulates the native liver module architecture and could be used for various applications such as early drug screening and disease modeling.


Author(s):  
D.J.P. Ferguson ◽  
A.R. Berendt ◽  
J. Tansey ◽  
K. Marsh ◽  
C.I. Newbold

In human malaria, the most serious clinical manifestation is cerebral malaria (CM) due to infection with Plasmodium falciparum. The pathology of CM is thought to relate to the fact that red blood cells containing mature forms of the parasite (PRBC) cytoadhere or sequester to post capillary venules of various tissues including the brain. This in vivo phenomenon has been studied in vitro by examining the cytoadherence of PRBCs to various cell types and purified proteins. To date, three Ijiost receptor molecules have been identified; CD36, ICAM-1 and thrombospondin. The specific changes in the PRBC membrane which mediate cytoadherence are less well understood, but they include the sub-membranous deposition of electron-dense material resulting in surface deformations called knobs. Knobs were thought to be essential for cytoadherence, lput recent work has shown that certain knob-negative (K-) lines can cytoadhere. In the present study, we have used electron microscopy to re-examine the interactions between K+ PRBCs and both C32 amelanotic melanoma cells and human umbilical vein endothelial cells (HUVEC).We confirm previous data demonstrating that C32 cells possess numerous microvilli which adhere to the PRBC, mainly via the knobs (Fig. 1). In contrast, the HUVEC were relatively smooth and the PRBCs appeared partially flattened onto the cell surface (Fig. 2). Furthermore, many of the PRBCs exhibited an invagination of the limiting membrane in the attachment zone, often containing a cytoplasmic process from the endothelial cell (Fig. 2).


Cells ◽  
2021 ◽  
Vol 10 (4) ◽  
pp. 876
Author(s):  
Raquel Bernad ◽  
Cian J. Lynch ◽  
Rocio G. Urdinguio ◽  
Camille Stephan-Otto Attolini ◽  
Mario F. Fraga ◽  
...  

Pluripotent stem cells can be stabilized in vitro at different developmental states by the use of specific chemicals and soluble factors. The naïve and primed states are the best characterized pluripotency states. Naïve pluripotent stem cells (PSCs) correspond to the early pre-implantation blastocyst and, in mice, constitute the optimal starting state for subsequent developmental applications. However, the stabilization of human naïve PSCs remains challenging because, after short-term culture, most current methods result in karyotypic abnormalities, aberrant DNA methylation patterns, loss of imprinting and severely compromised developmental potency. We have recently developed a novel method to induce and stabilize naïve human PSCs that consists in the simple addition of a chemical inhibitor for the closely related CDK8 and CDK19 kinases (CDK8/19i). Long-term cultured CDK8/19i-naïve human PSCs preserve their normal karyotype and do not show widespread DNA demethylation. Here, we investigate the long-term stability of allele-specific methylation at imprinted loci and the differentiation potency of CDK8/19i-naïve human PSCs. We report that long-term cultured CDK8/19i-naïve human PSCs retain the imprinting profile of their parental primed cells, and imprints are further retained upon differentiation in the context of teratoma formation. We have also tested the capacity of long-term cultured CDK8/19i-naïve human PSCs to differentiate into primordial germ cell (PGC)-like cells (PGCLCs) and trophoblast stem cells (TSCs), two cell types that are accessible from the naïve state. Interestingly, long-term cultured CDK8/19i-naïve human PSCs differentiated into PGCLCs with a similar efficiency to their primed counterparts. Also, long-term cultured CDK8/19i-naïve human PSCs were able to differentiate into TSCs, a transition that was not possible for primed PSCs. We conclude that inhibition of CDK8/19 stabilizes human PSCs in a functional naïve state that preserves imprinting and potency over long-term culture.


2021 ◽  
Vol 22 (9) ◽  
pp. 4334
Author(s):  
Katrina Albert ◽  
Jonna Niskanen ◽  
Sara Kälvälä ◽  
Šárka Lehtonen

Induced pluripotent stem cells (iPSCs) are a self-renewable pool of cells derived from an organism’s somatic cells. These can then be programmed to other cell types, including neurons. Use of iPSCs in research has been two-fold as they have been used for human disease modelling as well as for the possibility to generate new therapies. Particularly in complex human diseases, such as neurodegenerative diseases, iPSCs can give advantages over traditional animal models in that they more accurately represent the human genome. Additionally, patient-derived cells can be modified using gene editing technology and further transplanted to the brain. Glial cells have recently become important avenues of research in the field of neurodegenerative diseases, for example, in Alzheimer’s disease and Parkinson’s disease. This review focuses on using glial cells (astrocytes, microglia, and oligodendrocytes) derived from human iPSCs in order to give a better understanding of how these cells contribute to neurodegenerative disease pathology. Using glia iPSCs in in vitro cell culture, cerebral organoids, and intracranial transplantation may give us future insight into both more accurate models and disease-modifying therapies.


2019 ◽  
Vol 9 (1) ◽  
Author(s):  
Mariko Moriyama ◽  
Shunya Sahara ◽  
Kaori Zaiki ◽  
Ayumi Ueno ◽  
Koichi Nakaoji ◽  
...  

AbstractWound healing is regulated by complex interactions between the keratinocytes and other cell types including fibroblasts. Recently, adipose-derived mesenchymal stromal/stem cells (ASCs) have been reported to influence wound healing positively via paracrine involvement. However, their roles in keratinocytes are still obscure. Therefore, investigation of the precise effects of ASCs on keratinocytes in an in vitro culture system is required. Our recent data indicate that the epidermal equivalents became thicker on a collagen vitrigel membrane co-cultured with human ASCs (hASCs). Co-culturing the human primary epidermal keratinocytes (HPEK) with hASCs on a collagen vitrigel membrane enhanced their abilities for cell proliferation and adhesion to the membrane but suppressed their differentiation suggesting that hASCs could maintain the undifferentiated status of HPEK. Contrarily, the effects of co-culture using polyethylene terephthalate or polycarbonate membranes for HPEK were completely opposite. These differences may depend on the protein permeability and/or structure of the membrane. Taken together, our data demonstrate that hASCs could be used as a substitute for fibroblasts in skin wound repair, aesthetic medicine, or tissue engineering. It is also important to note that a co-culture system using the collagen vitrigel membrane allows better understanding of the interactions between the keratinocytes and ASCs.


Blood ◽  
2006 ◽  
Vol 107 (4) ◽  
pp. 1265-1275 ◽  
Author(s):  
Abby L. Olsen ◽  
David L. Stachura ◽  
Mitchell J. Weiss

Embryonic stem (ES) cells exhibit the remarkable capacity to become virtually any differentiated tissue upon appropriate manipulation in culture, a property that has been beneficial for studies of hematopoiesis. Until recently, the majority of this work used murine ES cells for basic research to elucidate fundamental properties of blood-cell development and establish methods to derive specific mature lineages. Now, the advent of human ES cells sets the stage for more applied pursuits to generate transplantable cells for treating blood disorders. Current efforts are directed toward adapting in vitro hematopoietic differentiation methods developed for murine ES cells to human lines, identifying the key interspecies differences in biologic properties of ES cells, and generating ES cell-derived hematopoietic stem cells that are competent to repopulate adult hosts. The ultimate medical goal is to create patient-specific and generic ES cell lines that can be expanded in vitro, genetically altered, and differentiated into cell types that can be used to treat hematopoietic diseases.


Sign in / Sign up

Export Citation Format

Share Document