Summary

通过成熟的脂肪细胞在天花板文化去分化代人脂肪干细胞

Published: March 07, 2015
doi:

Summary

Mature adipocytes may represent an abundant source of stem cells through dedifferentiation, which leads to a homogenous population of fibroblast-like cells. Collagenase digestion is used to isolate mature adipocytes from human fat. The goal of our protocol is to obtain multipotent, dedifferentiated fat cells from human mature adipocytes.

Abstract

Mature adipocytes have been shown to reverse their phenotype into fibroblast-like cells in vitro through a technique called ceiling culture. Mature adipocytes can also be isolated from fresh adipose tissue for depot-specific characterization of their function and metabolic properties. Here, we describe a well-established protocol to isolate mature adipocytes from adipose tissues using collagenase digestion, and subsequent steps to perform ceiling cultures. Briefly, adipose tissues are incubated in a Krebs-Ringer-Henseleit buffer containing collagenase to disrupt tissue matrix. Floating mature adipocytes are collected on the top surface of the buffer. Mature cells are plated in a T25-flask completely filled with media and incubated upside down for a week. An alternative 6-well plate culture approach allows the characterization of adipocytes undergoing dedifferentiation. Adipocyte morphology drastically changes over time of culture. Immunofluorescence can be easily performed on slides cultivated in 6-well plates as demonstrated by FABP4 immunofluorescence staining. FABP4 protein is present in mature adipocytes but down-regulated through dedifferentiation of fat cells. Mature adipocyte dedifferentiation may represent a new avenue for cell therapy and tissue engineering.

Introduction

In vitro dedifferentiation of mature adipocytes is achieved through a technique called ceiling culture1. Because of their natural tendency to float in aqueous solutions, isolated mature adipocytes adhere to the surface of an inverted flask fully filled with culture medium. Over a few days, cells modify their spherical morphology and become fibroblast-like cells. The resulting cells, called dedifferentiated fat (DFAT) cells, are multipotent2. Research articles on adipocyte dedifferentiation, especially on human cells, are limited. However, they have already provided interesting information regarding multipotency, cell phenotype and replicative capacity of DFAT cells2. Mature adipocytes originating from various fat compartments have been successfully dedifferentiated including those originating from human visceral and subcutaneous adipose tissues2-4. In addition to these depots, Kishimoto and collaborators sampled adipose tissue from the buccal fat pads and dedifferentiated adipocytes into DFAT cells5. Matsumoto and collaborators successfully generated subcutaneous DFAT cells from patients covering a wide range of ages, and the majority of cells had a high proliferative rate and less than 6% of senescence even after 10 passages in culture2.

DFAT cells have been successfully re-differentiated into several lineages, including adipogenic, osteogenic, chondrogenic and neurogenic lineages2,3,6. These cells express several embryonic stem cell markers such as Nanog and the four identified pluripotent factors Oct4, c-myc, Klf4 and Sox23. They also express markers specific to each of the three germ layers7. In addition, DFAT cells are similar to Bone Marrow-derived Mesenchymal Stem Cells (BM-derived MSC) based on their epigenetic signature3. Exploiting the stem cell capacity of DFAT cells, many groups have investigated their potential to treat or improve various diseases8,9. Improvements of pathologic conditions, such as infracted cardiac tissue, spinal cord injury and urethral sphincter dysfunction, have been observed with DFAT cell injections in rat models of disease10-12.

In addition to the stem cell properties of DFAT cells, they may represent a new cellular model for adipocyte physiology studies. The 3T3-L1 cell line is often used for this purpose as these cells differentiate into adherent, lipid-storing adipocytes under adipogenic stimulation13. However, these cells originate from mouse embryo tissue13. Also, depot-specificity cannot be investigated with this model and it may not fully reflect human adipocyte physiology14. Other laboratories work with isolated adipose cells from murine fat depots, but fat distribution is not dimorphic in mice and anatomical configuration of the rodent’s abdominal cavity prevents from extrapolating directly to humans15. In order to study adipocytes in the context of the physiopathology of human obesity, consideration of body fat distribution and fat depot-specific differences has become essential16. Some limitations of primary preadipocyte cultures, including cell quantities obtained from adipose tissue biopsy samples and their senescence after a few passages in culture, created the need for alternate models. Perrini and collaborators investigated depot-specificity in gene expression of DFAT cells originating from visceral and subcutaneous fat and compared them to adipose-derived stem cells (ASC) from the same fat depot. They demonstrated that differences in gene expression and function where mainly found between depots than between cell types, suggesting that DFAT cells are physiologically close to ASC from the same depot. DFAT cells may represent an interesting alternative to available models for studies on fat distribution in the pathophysiology of human obesity. Moreover, ceiling culture is a promising method to obtain adult stem cells for tissue engineering purposes.

Here, we describe collagenase digestion, a widely-used technique to isolate mature adipocytes from the subcutaneous and/or visceral fat samples17, and the subsequent steps to perform ceiling culture and dedifferentiate these cells into multipotent, fibroblast-like cells.

Protocol

伦理学声明:该项目已通过IUCPQ的研究伦理委员会之前,患者招募。在这篇文章中/视频的目的,我们从2例患者获得组织:1)一名62岁的男性患者,50.7公斤/米2和2)35岁的女性患者体重指数与57公斤体重/米2。实验可以与脂肪车厢做,但都被限制在一个隔脂肪这部影片的目的。视频技术方面进行与患者1,并与去分化细胞来自患者2进行FABP4免疫。 1.样品处理<…

Representative Results

主要形态变化发生去分化( 图1)中成熟脂肪细胞。 如图2中 ,细胞经历去分化染色用抗FABP4抗体进行荧光分析。细胞与圆形形态表达的FABP4蛋白,而大多数的成纤维细胞样细胞的没有。去分化后,DFAT细胞可以培养使用标准程序几个传代。我们已经能够达到超过15代对人网膜和皮下DFAT细胞系(数据未示出)。 <img alt="图1" src…

Discussion

成熟的脂肪细胞与天花板培养技术去分化是一种新的方法来从本地脂肪组织的一个小样本的获取脂肪干细胞。根据我们的经验和其他人2,一克组织足以在板25-cm 2的烧瓶中并以获得DFAT细胞的量的均匀性已被证实由波洛尼和合作者3群。脂肪细胞分化似乎独立他们的年龄,性别和其他特征可以与来自任何供体细胞。间所产生的DFAT获得人口,仍然存在一些圆形或部分细长的细?…

Divulgazioni

The authors have nothing to disclose.

Acknowledgements

This study was supported by Natural Sciences and Engineering Research Council of Canada Discovery Grant (371697-2011, AT). The authors want to acknowledge the help of bariatric surgeons Drs S. Biron, F-S. Hould, S. Lebel, O. Lescelleur, P. Marceau as well as Christine Racine and Caroline Gagnon from the IUCPQ Tissue Bank. We thank Mr Jacques Cadorette from the IUCPQ’s audiovisual services for video shooting and editing.

Materials

Bovine serum albumine Sigma A7906
Adenosine Sigma A4036
Ascorbic acid Sigma A0278
NaCl Any brand can be used
KCl Any brand can be used
CaCl2 Any brand can be used
MgCl2 Any brand can be used
KH2PO4 Any brand can be used
HEPES Any brand can be used
Glucose Any brand can be used
Type I collagenase Worthington Biochemical Corp LS-004196
DMEM/F-12, HEPES, no phenol red Gibco-Life Technologies 11039-021 Add to medium : 20% calf serum, gentamicin (50µg/ml) and fungizone (2.5µg/ml)
Calf Serum, iron supplemented, from formula-fed calves Sigma C8056-500ml
1/2 In plastic bushing Iberville 2704-CP SKU:1000120918 (Home Depot)
Liquid nitrogen Linde
Formalin soluton, neutral buffered, 10% SIGMA HT501128
Sterile tweezers
Sterile scissors
60cc syringes BD Syringe
Plastic tubing
Krebs-Ringer-Henseleit stock buffer (KRH) Prepare stock buffer as following: 25mM HEPES pH7.6, 125mM NaCl, 3.73mM KCl, 5mM CaCl2.2H2O, 2.5mM MgCl2.6H2O, 1mM K2HPO4. Adjust pH to 7.4.
Krebs-Ringer-Henseleit-Working Buffer (KRH-WB) Add the following components freshly to KRH buffer: 4% bovine serum albumin, 5mM glucose, 0.1µM adenosine, 560 µM ascorbic acid
KRH-WB supplemented with Type I collagenase Add 350U/ml of Type I collagenase
T25 unvented cap tissue culture flask Sarsted or other brand
6-well tissue culture plate BD Falcon or other brand
Microscope cover glass 22×22 Fisherbrand 12-542-B
Sterile beakers

Riferimenti

  1. Zhang, H. H., Kumar, S., Barnett, A. H., Eggo, M. C. Ceiling culture of mature human adipocytes: use in studies of adipocyte functions. J Endocrinol. 164 (2), 119-128 (2000).
  2. Matsumoto, T., et al. Mature adipocyte-derived dedifferentiated fat cells exhibit multilineage potential. J Cell Physiol. 215 (1), 210-222 (2008).
  3. Poloni, A., et al. Human dedifferentiated adipocytes show similar properties to bone marrow-derived mesenchymal stem cells. Stem Cells. 30 (5), 965-974 (2012).
  4. Perrini, S., et al. Differences in gene expression and cytokine release profiles highlight the heterogeneity of distinct subsets of adipose tissue-derived stem cells in the subcutaneous and visceral adipose tissue in humans. PLoS One. 8 (3), e57892 (2013).
  5. Kishimoto, N., et al. The osteoblastic differentiation ability of human dedifferentiated fat cells is higher than that of adipose stem cells from the buccal fat pad. Clin Oral Investig. , (2013).
  6. Kou, L., et al. The phenotype and tissue-specific nature of multipotent cells derived from human mature adipocytes. Biochem Biophys Res Commun. 444 (4), 543-548 (2014).
  7. Jumabay, M., et al. Pluripotent stem cells derived from mouse and human white mature adipocytes. Stem Cells Transl Med. 3 (2), 161-171 (2014).
  8. Sugawara, A., Sato, S. Application of dedifferentiated fat cells for periodontal tissue regeneration. Hum Cell. 27 (1), 12-21 (2014).
  9. Kikuta, S., et al. Osteogenic effects of dedifferentiated fat cell transplantation in rabbit models of bone defect and ovariectomy-induced osteoporosis. Tissue Eng Part A. 19 (15-16), 1792-1802 (2013).
  10. Obinata, D., et al. Transplantation of mature adipocyte-derived dedifferentiated fat (DFAT) cells improves urethral sphincter contractility in a rat model. Int J Urol. 18 (12), 827-834 (2011).
  11. Jumabay, M., et al. Dedifferentiated fat cells convert to cardiomyocyte phenotype and repair infarcted cardiac tissue in rats. J Mol Cell Cardiol. 47 (5), 565-575 (2009).
  12. Ohta, Y., et al. Mature adipocyte-derived cells, dedifferentiated fat cells (DFAT), promoted functional recovery from spinal cord injury-induced motor dysfunction in rats. Cell Transplant. 17 (8), 877-886 (2008).
  13. Moreno-Navarrete, J. M. F. -. r., Symonds, M. E. Ch. 2. Adipose Tissue Biology. , 17-38 (2012).
  14. Poulos, S. P., Dodson, M. V., Hausman, G. J. Cell line models for differentiation: preadipocytes and adipocytes. Exp Biol Med (Maywood. 235 (10), 1185-1193 (2010).
  15. Casteilla, L., Penicaud, L., Cousin, B., Calise, D. Choosing an adipose tissue depot for sampling: factors in selection and depot specificity). Methods Mol Biol. 456, 23-38 (2008).
  16. Tchernof, A., Despres, J. P. Pathophysiology of human visceral obesity: an update. Physiol Rev. 93 (1), 359-404 (2013).
  17. Rodbell, M. Metabolism of Isolated Fat Cells. I. Effects of Hormones on Glucose Metabolism and Lipolysis. J Biol Chem. 239, 375-380 (1964).
  18. Watson, J. E., et al. Comparison of Markers and Functional Attributes of Human Adipose-Derived Stem Cells and Dedifferentiated Adipocyte Cells from Subcutaneous Fat of an Obese Diabetic Donor. Adv Wound Care. 3 (3), 219-228 (2014).
check_url/it/52485?article_type=t

Play Video

Citazione di questo articolo
Lessard, J., Côté, J. A., Lapointe, M., Pelletier, M., Nadeau, M., Marceau, S., Biertho, L., Tchernof, A. Generation of Human Adipose Stem Cells through Dedifferentiation of Mature Adipocytes in Ceiling Cultures. J. Vis. Exp. (97), e52485, doi:10.3791/52485 (2015).

View Video