1
|
Che Y, Shimizu Y, Murohara T. Therapeutic Potential of Adipose-Derived Regenerative Cells for Ischemic Diseases. Cells 2025; 14:343. [PMID: 40072072 PMCID: PMC11898683 DOI: 10.3390/cells14050343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/21/2025] [Accepted: 02/26/2025] [Indexed: 03/15/2025] Open
Abstract
Adipose-derived regenerative cells (ADRCs) are one of the most promising cell sources that possess significant therapeutic effects. They have now become a main source of cell therapy for the treatment of ischemic diseases due to their easy accessibility, expansion, and differentiation. Additionally, ADRCs can release multiple paracrine factors and extracellular vesicles that contribute to tissue regeneration by promoting angiogenesis, regulating inflammation, alleviating apoptosis, and inhibiting fibrosis. However, ADRCs still have some limitations to realize their full therapeutic potential. To address these issues, protective mechanistic studies and bioengineering studies have been carried out. This review focused on the recently studied mechanisms, such as paracrine factors, cell fusion, and mitochondrial transfer, involving the therapeutic potential of ADRCs in ischemic diseases and discussed some modification techniques of ADRCs.
Collapse
Affiliation(s)
| | - Yuuki Shimizu
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | | |
Collapse
|
2
|
Garroni G, Cruciani S, Serra D, Pala R, Coradduzza D, Cossu ML, Ginesu GC, Ventura C, Maioli M. Effects of the MCF-7 Exhausted Medium on hADSC Behaviour. Int J Mol Sci 2024; 25:7026. [PMID: 39000134 PMCID: PMC11241546 DOI: 10.3390/ijms25137026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/19/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Stem cells possess the ability to differentiate into different lineages and the ability to self-renew, thus representing an excellent tool for regenerative medicine. They can be isolated from different tissues, including the adipose tissue. Adipose tissue and human adipose-derived stem cells (hADSCs) are privileged candidates for regenerative medicine procedures or other plastic reconstructive surgeries. The cellular environment is able to influence the fate of stem cells residing in the tissue. In a previous study, we exposed hADSCs to an exhausted medium of a breast cancer cell line (MCF-7) recovered at different days (4, 7, and 10 days). In the same paper, we inferred that the medium was able to influence the behaviour of stem cells. Considering these results, in the present study, we evaluated the expression of the major genes related to adipogenic and osteogenic differentiation. To confirm the gene expression data, oil red and alizarin red colorimetric assays were performed. Lastly, we evaluated the expression of miRNAs influencing the differentiation process and the proliferation rate, maintaining a proliferative state. The data obtained confirmed that cells exposed to the medium maintained a stem and proliferative state that could lead to a risky proliferative phenotype.
Collapse
Affiliation(s)
- Giuseppe Garroni
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (G.G.); (S.C.); (D.S.); (R.P.); (D.C.)
| | - Sara Cruciani
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (G.G.); (S.C.); (D.S.); (R.P.); (D.C.)
| | - Diletta Serra
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (G.G.); (S.C.); (D.S.); (R.P.); (D.C.)
| | - Renzo Pala
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (G.G.); (S.C.); (D.S.); (R.P.); (D.C.)
| | - Donatella Coradduzza
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (G.G.); (S.C.); (D.S.); (R.P.); (D.C.)
| | - Maria Laura Cossu
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy; (M.L.C.); (G.C.G.)
| | - Giorgio Carlo Ginesu
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy; (M.L.C.); (G.C.G.)
| | - Carlo Ventura
- National Laboratory of Molecular Biology and Stem Cell Bioengineering of the National Institute of Biostructures and Biosystems (NIBB) c/o Eldor Lab, Via Corticella 183, 40129 Bologna, Italy;
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (G.G.); (S.C.); (D.S.); (R.P.); (D.C.)
- Center for Developmental Biology and Reprogramming (CEDEBIOR), Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy
| |
Collapse
|
3
|
Dong L, Li X, Leng W, Guo Z, Cai T, Ji X, Xu C, Zhu Z, Lin J. Adipose stem cells in tissue regeneration and repair: From bench to bedside. Regen Ther 2023; 24:547-560. [PMID: 37854632 PMCID: PMC10579872 DOI: 10.1016/j.reth.2023.09.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/21/2023] [Accepted: 09/28/2023] [Indexed: 10/20/2023] Open
Abstract
ADSCs are a large number of mesenchymal stem cells in Adipose tissue, which can be applied to tissue engineering. ADSCs have the potential of multi-directional differentiation, and can differentiate into bone tissue, cardiac tissue, urothelial cells, skin tissue, etc. Compared with other mesenchymal stem cells, ADSCs have a multitude of promising advantages, such as abundant number, accessibility in cell culture, stable function, and less immune rejection. There are two main methods to use ADSCs for tissue repair and regeneration. One is to implant the "ADSCs-scaffold composite" into the injured site to promote tissue regeneration. The other is cell-free therapy: using ADSC-exos or ADSC-CM alone to release a large number of miRNAs, cytokines and other bioactive substances to promote tissue regeneration. The tissue regeneration potential of ADSCs is regulated by a variety of cytokines, signaling molecules, and external environment. The differentiation of ADSCs into different tissues is also induced by growth factors, ions, hormones, scaffold materials, physical stimulation, and other factors. The specific mechanisms are complex, and most of the signaling pathways need to be further explored. This article reviews and summarizes the mechanism and clinical application of ADSCs in tissue injury repair so far, and puts forward further problems that need to be solved in this field, hoping to provide directions for further research in this field.
Collapse
Affiliation(s)
- Lei Dong
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Xiaoyu Li
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Wenyuan Leng
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Zhenke Guo
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Tianyu Cai
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Xing Ji
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Chunru Xu
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Zhenpeng Zhu
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Jian Lin
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| |
Collapse
|
4
|
Jovic D, Preradovic L, Kremenovic M, Jovic F, Antonic M, Aleksic Z, Ljubojevic V. Effect of Donor Site Selection for Fat Grafting on the Yield and Viability of the Stromal Vascular Fraction. Aesthet Surg J 2023; 43:NP704-NP712. [PMID: 37289983 DOI: 10.1093/asj/sjad184] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/25/2023] [Accepted: 05/30/2023] [Indexed: 06/10/2023] Open
Abstract
BACKGROUND The efficacy of stromal vascular fraction (SVF) treatment, or stem cell treatment, directly depends on the SVF cell count and the cells' viability. The SVF cell count and viability are in direct correlation with the adipose tissue harvesting site that yields SVF cells, making this research a contribution to developing tissue guidance. OBJECTIVES The aim of this study was to investigate the importance of harvesting subcutaneous adipose tissue-derived SVF cells on the concentration and viability of SVF. METHODS Adipose tissue was collected by vibration-assisted liposuction from the regions of the upper and lower abdomen, lumbar region, and inner thigh region. With the semiautomatic UNISTATION 2nd Version system, the obtained fat was chemically processed (with collagenase enzyme) and a concentrate of SVF cells was obtained by centrifugation. These samples were then analyzed with the Luna-Stem Counter device to measure the number and viability of SVF cells. RESULTS When comparing the regions of the upper abdomen, lower abdomen, lumbar region, and inner thigh, the highest concentration of SVF was found in the lumbar region, specifically at an average of 97,498.00 per 1.0 mL of concentrate. The lowest concentration was found in the upper abdominal region. When ranking the viability values, the highest cell viability of SVF was observed in the lumbar region, measuring 36.6200%. The lowest viability was found in the upper abdominal region, measuring 24.4967%. CONCLUSIONS By comparing the upper and lower abdominal, lumbar, and inner thigh regions, the authors have come to the conclusion that, on average, the largest number of cells with the highest viability was obtained from the lumbar region.
Collapse
|
5
|
Hamel KM, Liimatta KQ, Belgodere JA, Bunnell BA, Gimble JM, Martin EC. Adipose-Derived Stromal/Stem Cell Response to Tumors and Wounds: Evaluation of Patient Age. Stem Cells Dev 2022; 31:579-592. [PMID: 35262397 PMCID: PMC9836707 DOI: 10.1089/scd.2021.0280] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 03/05/2022] [Indexed: 01/22/2023] Open
Abstract
Tumors were characterized as nonhealing wounds by Virchow in 1858 and Dvorak in 1986. Since then, researchers have analyzed tumors from a new perspective. The parallels between tumorigenesis and physiological wound healing can provide a new framework for developing antitumor therapeutics. One commonality between tumors and wounds is the involvement of the stromal environment, particularly adipose stromal/stem cells (ASCs). ASCs exhibit dual functions, in which they stimulate tumor progression and assist in tissue repair and regeneration. Numerous studies have focused on the role of ASCs in cancer and wound healing, but none to date has linked age, cancer, and wound healing. Furthermore, very few studies have focused on the role of donor-specific characteristics of ASCs, such as age and their role in facilitating ASC behavior in cancer and wound healing. This review article is designed to provide important insights into the impact of donor age on ASC tumor and wound response and their role in facilitating ASC behavior in cancer and wound healing.
Collapse
Affiliation(s)
- Katie M. Hamel
- Department of Biological Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Kara Q. Liimatta
- Department of Biological Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Jorge A. Belgodere
- Department of Biological Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Bruce A. Bunnell
- University of North Texas Health Sciences Center, Fort Worth, Texas, USA
| | | | - Elizabeth C. Martin
- Department of Biological Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| |
Collapse
|
6
|
Kempa S, Brix E, Heine N, Hösl V, Strauss C, Eigenberger A, Brébant V, Seitz S, Prantl L. Autologous fat grafting for breast reconstruction after breast cancer: a 12-year experience. Arch Gynecol Obstet 2021; 305:921-927. [PMID: 34532758 PMCID: PMC8967754 DOI: 10.1007/s00404-021-06241-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/07/2021] [Indexed: 12/24/2022]
Abstract
Purpose The aim of our study was to examine the surgical outcome and complications (efficiency) as well as the incidence of locoregional recurrence and distant metastases (oncological safety) in patients who underwent autologous fat grafting (AFG) of the breast following breast cancer surgery. Methods In our monocentric cohort study, retrospective and prospective data were collected from all consecutive patients who underwent AFG after breast cancer between 2008 and 2020; a total of 93 patients met the inclusion criteria. Results Our long-term results showed no increase in tumor recurrence and distant metastases in the studied collective when compared to the available literature. We observed 1 local recurrence (1.1%), 2 distant metastases (2.2%), and 1 tumor-related death (1.1%). There was a high degree of patient satisfaction; 67.12% of patients reported adequate satisfaction with autologous fat grafting. Conclusion Currently, to our knowledge, this is the study with the longest follow-up time (mean 6.7 years after AFG and 11.5 years after tumor resection). The results of our clinical study will contribute to improve evidence in the broad field of AFG, adipose stem cell and tumor research. Consistent with our study, the literature review shows a clear tendency of clinical trial results with a low incidence rate of tumor recurrence and metastasis following the use of AFG. AFG seems to be a safe procedure also after breast cancer treatment.
Collapse
Affiliation(s)
- Sally Kempa
- Department of Plastic, Aesthetic and Reconstructive Surgery, University of Regensburg, Franz-Josef-Strauss-Allee 11, D-93053, Regensburg, Germany
| | - Eva Brix
- Department of Plastic, Aesthetic and Reconstructive Surgery, University of Regensburg, Franz-Josef-Strauss-Allee 11, D-93053, Regensburg, Germany
| | - Norbert Heine
- Department of Plastic, Aesthetic and Reconstructive Surgery, University of Regensburg, Franz-Josef-Strauss-Allee 11, D-93053, Regensburg, Germany
| | - Vanessa Hösl
- Department of Plastic, Aesthetic and Reconstructive Surgery, University of Regensburg, Franz-Josef-Strauss-Allee 11, D-93053, Regensburg, Germany
| | - Catharina Strauss
- Department of Plastic, Aesthetic and Reconstructive Surgery, University of Regensburg, Franz-Josef-Strauss-Allee 11, D-93053, Regensburg, Germany
| | - Andreas Eigenberger
- Department of Plastic, Aesthetic and Reconstructive Surgery, University of Regensburg, Franz-Josef-Strauss-Allee 11, D-93053, Regensburg, Germany
| | - Vanessa Brébant
- Department of Plastic, Aesthetic and Reconstructive Surgery, University of Regensburg, Franz-Josef-Strauss-Allee 11, D-93053, Regensburg, Germany
| | - Stephan Seitz
- Department of Obstetrics and Gynecology, Caritas Hospital St. Josef, University of Regensburg, D-93053, Regensburg, Germany
| | - Lukas Prantl
- Department of Plastic, Aesthetic and Reconstructive Surgery, University of Regensburg, Franz-Josef-Strauss-Allee 11, D-93053, Regensburg, Germany.
| |
Collapse
|
7
|
Abstract
Scar is a common way of healing after tissue injury. The poor scar healing will not only cause dysfunction of tissues and organs but also affect the appearance of the patients’ body surface, which causes the pressure of life and spirit to the patients. However, the formation of scar tissue is an extremely complex process and its mechanism is not fully understood. At present, there is no treatment method to eliminate scars completely. Fibroblasts are the most abundant cells in the dermis, which have the ability to synthesize and remodel extracellular matrix (ECM). Myofibroblasts actively participate in the wound healing process and influence the outcome. Therefore, both of them play important roles in wound healing and scar formation. Adipose tissue-derived stem cells (ADSCs) are pluripotent stem cells that can act on target cells by paracrine. Adipose tissue stem cell-derived exosomes (ADSC-Exos) are important secretory substances of ADSCs. They are nanomembrane vesicles that can transport a variety of cellular components and fuse with target cells. In this review, we will discuss the effects of ADSCs and ADSC-Exos on the behavior of fibroblasts and myofibroblasts during wound healing and scarring stage in combination with recent studies.
Collapse
|
8
|
Rafiei Alavi SN, Madani Neishaboori A, Hossein H, Sarveazad A, Yousefifard M. Efficacy of adipose tissue-derived stem cells in locomotion recovery after spinal cord injury: a systematic review and meta-analysis on animal studies. Syst Rev 2021; 10:213. [PMID: 34330329 PMCID: PMC8325264 DOI: 10.1186/s13643-021-01771-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 07/21/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Considerable disparities exist on the use of adipose tissue-derived stem cells (ADSCs) for treatment of spinal cord injury (SCI). Hence, the current systematic review aimed to investigate the efficacy of ADSCs in locomotion recovery following SCI in animal models. METHODS A search was conducted in electronic databases of MEDLINE, Embase, Scopus, and Web of Science until the end of July 2019. Reference and citation tracking and searching Google and Google Scholar search engines were performed to achieve more studies. Animal studies conducted on rats having SCI which were treated with ADSCs were included in the study. Exclusion criteria were lacking a non-treated control group, not evaluating locomotion, non-rat studies, not reporting the number of transplanted cells, not reporting isolation and preparation methods of stem cells, review articles, combination therapy, use of genetically modified ADSCs, use of induced pluripotent ADSCs, and human trials. Risk of bias was assessed using Hasannejad et al.'s proposed method for quality control of SCI-animal studies. Data were analyzed in STATA 14.0 software, and based on a random effect model, pooled standardized mean difference with a 95% confidence interval was presented. RESULTS Of 588 non-duplicated papers, data from 18 articles were included. Overall risk of bias was high risk in 8 studies, some concern in 9 studies and low risk in 1 study. Current evidence demonstrated that ADSCs transplantation could improve locomotion following SCI (standardized mean difference = 1.71; 95%CI 1.29-2.13; p < 0.0001). A considerable heterogeneity was observed between the studies (I2 = 72.0%; p < 0.0001). Subgroup analysis and meta-regression revealed that most of the factors like injury model, the severity of SCI, treatment phase, injury location, and number of transplanted cells did not have a significant effect on the efficacy of ADSCs in improving locomotion following SCI (pfor odds ratios > 0.05). CONCLUSION We conclude that any number of ADSCs by any prescription routes can improve locomotion recovery in an SCI animal model, at any phase of SCI, with any severity. Given the remarkable bias about blinding, clinical translation of the present results is tough, because in addition to the complexity of the nervous system and the involvement of far more complex motor circuits in the human, blinding compliance and motor outcome assessment tests in animal studies and clinical trials are significantly different.
Collapse
Affiliation(s)
| | - Arian Madani Neishaboori
- Physiology Research Center, Iran University of Medical Sciences, Hemmat Highway, P.O Box: 14665-354, Tehran, Iran
| | - Hasti Hossein
- Physiology Research Center, Iran University of Medical Sciences, Hemmat Highway, P.O Box: 14665-354, Tehran, Iran
| | - Arash Sarveazad
- Colorectal Research Center, Iran University of Medical Sciences, Niayesh St, Satarkhan Av, P.O Box: 14665-354, 1449614535, Tehran, Iran. .,Nursing Care Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Mahmoud Yousefifard
- Physiology Research Center, Iran University of Medical Sciences, Hemmat Highway, P.O Box: 14665-354, Tehran, Iran.
| |
Collapse
|
9
|
Nguyen Thanh L, Dam PTM, Nguyen HP, Nguyen TST, To HM, Nguyen HB, Luu NA, Hoang DM. Can Autologous Adipose-Derived Mesenchymal Stem Cell Transplantation Improve Sexual Function in People with Sexual Functional Deficiency? Stem Cell Rev Rep 2021; 17:2153-2163. [PMID: 34129158 DOI: 10.1007/s12015-021-10196-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Sexual functional deficiency occurs at some point in life and becomes a problematic issue in middle-aged adulthood. Regenerative medicine, especially mesenchymal stem cell (MSC) transplantation, has developed extensively, with preclinical and clinical trials emphasizing the benefits of stem cell therapy for restoration of sexual deficiency. This study was designed to develop a new therapeutic stem cell treatment for people with sexual functional deficiency. METHODS Thirty-one patients, including 15 males and 16 females with a medical history of reduced sexual activity, met the inclusion criteria and were enrolled in the study, phase I/IIa clinical trial with a 12-month follow-up. Adipose tissue-derived mesenchymal stem/stromal cells (ADSC) were isolated by type I collagenase digestion and cultured at the Stem Cell Core Facility under ISO 14644-1. Each participant received 1 million cells/kg of body weight via the intravenous route. Safety was evaluated by assessing the occurrence of adverse events or severe adverse events. Efficacy was assessed in males by monitoring testosterone levels and administering the International Index of Erectile Function (IIEF) questionnaire and in females by monitoring anti-Mullerian hormone (AMH), estradiol (E2), and follicle-stimulating hormone (FSH) levels and administering the Female Sexual Functioning Index (FSFI) questionnaire at baseline and 3-, 6-, and 12-months post-transplantation. RESULTS There was no occurrence of severe adverse events after ADSC administration in our study. Post-transplantation sexual satisfaction was observed in all patients enrolled in this study. Testosterone levels in males increased soon after transplantation and were maintained at high levels for up to 6 months before decreasing again at the 12-month follow-up. No significant changes in AMH, FSH or E2 levels were recorded in female patients. CONCLUSIONS Autologous ADSC infusion is a potential therapeutic option for patients with reduced sexual activity, especially for male patients. TRIAL REGISTRATION ClinicalTrials.gov. NCT03346967, Registered November 20, 2017.
Collapse
Affiliation(s)
- Liem Nguyen Thanh
- Vinmec Research Institute of Stem Cell and Gene Technology (VRISG), Vinmec Health Care System, Hanoi, Vietnam.
| | - Phuong T M Dam
- Vinmec Hightech Center, Vinmec Healthcare System, Hanoi, Vietnam
| | - Hoang-Phuong Nguyen
- Vinmec Research Institute of Stem Cell and Gene Technology (VRISG), Vinmec Health Care System, Hanoi, Vietnam
| | - Tan-Sinh Thi Nguyen
- Vinmec Times City International Hospital, 458 Minh Khai Street, Hanoi, Vietnam
| | - Huong Minh To
- Vinmec Times City International Hospital, 458 Minh Khai Street, Hanoi, Vietnam
| | - Hung Ba Nguyen
- Vinmec Times City International Hospital, 458 Minh Khai Street, Hanoi, Vietnam
| | - Ngoc-Anh Luu
- Vinmec Research Institute of Stem Cell and Gene Technology (VRISG), Vinmec Health Care System, Hanoi, Vietnam
| | - Duc M Hoang
- Vinmec Research Institute of Stem Cell and Gene Technology (VRISG), Vinmec Health Care System, Hanoi, Vietnam
| |
Collapse
|
10
|
Karimi R, Barabadi Z, Larijani B, Tavoosidana G, Lotfibakhshaiesh N, Absalan M, Jabbarpour Z, Ostad SN, Ai J. Comparison of insulin secretion by transduced adipose-derived and endometrial-derived stem cells in 2D and 3D cultures on fibrin scaffold. J Biomed Mater Res A 2021; 109:1036-1044. [PMID: 32862549 DOI: 10.1002/jbm.a.37094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 12/16/2022]
Abstract
Type 1 diabetes is a metabolic disorder caused by the loss or dysfunction of β-cells in the pancreas. Organ shortage is a critical concern of diabetic patients in need of beta islet transplantation. Tissue engineered islets are promising alternatives to traditional organ transplantation. Recent progress in stem cell biology and gene cloning techniques has raised hopes for the generation of insulin producing cells (IPCs) without the need of immunosuppression. The purpose of this study was to produce IPCs using human adipose-derived stem cells (hADSCs) and human endometrial-derived stem cells (hEnSCs) and also to compare the level of insulin secretion by these cells in 2D and 3D culture systems on fibrin scaffolding. Stem cells differentiation was carried out through transduction with an insulin over expression lentiviral vector. Real-time PCR and immunocytochemistry confirmed the successful transduction of both cell types. Both cell types showed comparable insulin secretion by ELISA.3D culture resulted in higher amounts of insulin secretion of the two cell types versus 2D as control. This study showed that insulin gene delivery to the stem cells could be an efficient method for producing IPCs and fibrin encapsulation enhances the functionality of these cells.
Collapse
Affiliation(s)
- Roya Karimi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Barabadi
- Department of Tissue Engineering, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Tavoosidana
- Department of Molecular medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasrin Lotfibakhshaiesh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Moloud Absalan
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Jabbarpour
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Naser Ostad
- Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Da Silva D, Crous A, Abrahamse H. Photobiomodulation: An Effective Approach to Enhance Proliferation and Differentiation of Adipose-Derived Stem Cells into Osteoblasts. Stem Cells Int 2021; 2021:8843179. [PMID: 33833810 PMCID: PMC8012132 DOI: 10.1155/2021/8843179] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 03/07/2021] [Accepted: 03/11/2021] [Indexed: 01/07/2023] Open
Abstract
Osteoporosis is regarded as the most common chronic metabolic bone condition in humans. In osteoporosis, bone mesenchymal stem cells (MSCs) have reduced cellular function. Regenerative medicine using adipose-derived stem cell (ADSC) transplantation can promote the growth and strength of new bones, improve bone stability, and reduce the risk of fractures. Various methods have been attempted to differentiate ADSCs to functioning specialized cells for prospective clinical application. However, commonly used therapies have resulted in damage to the donor site and morbidity, immune reactions, carcinogenic generation, and postoperative difficulties. Photobiomodulation (PBM) improves ADSC differentiation and proliferation along with reducing clinical difficulties such as treatment failures to common drug therapies and late initiation of treatment. PBM is a noninvasive, nonthermal treatment that encourages cells to produce more energy and to undergo self-repair by using visible green and red and invisible near-infrared (NIR) radiation. The use of PBM for ADSC proliferation and differentiation has been widely studied with multiple outcomes observed due to laser fluence and wavelength dependence. In this article, the potential for differentiating ADSCs into osteoblasts and the various methods used, including biological induction, chemical induction, and PBM, will be addressed. Likewise, the optimal laser parameters that could improve the proliferation and differentiation of ADSC, translating into clinical success, will be commented on.
Collapse
Affiliation(s)
- Daniella Da Silva
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein, Johannesburg, South Africa 2028
| | - Anine Crous
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein, Johannesburg, South Africa 2028
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein, Johannesburg, South Africa 2028
| |
Collapse
|
12
|
Anti-Oxidative Effects of Human Adipose Stem Cell Conditioned Medium with Different Basal Medium during Mouse Embryo In Vitro Culture. Animals (Basel) 2020; 10:ani10081414. [PMID: 32823702 PMCID: PMC7459530 DOI: 10.3390/ani10081414] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/02/2020] [Accepted: 08/04/2020] [Indexed: 01/20/2023] Open
Abstract
Simple Summary Assisted reproductive techniques, which are used to resolve various infertility problems, have advanced following the emphasis on their use. Embryos produced in vitro rather than in vivo are exposed to greater stress, with the quality of the embryos being affected by the in vitro culture conditions. To reduce oxidative stress and consequent apoptosis of embryos for successful implantation and pregnancy maintenance, the present study evaluated the anti-oxidative effect of human adipose stem cell conditioned medium (ASC-CM) with different basal medium as supplement in in vitro culture (IVC) medium for mouse preimplantation embryo. Treatment of 5% human ASC-CM based on Dulbecco′s modified Eagle′s medium (DMEM-CM) indicated an enhanced development of mouse in vitro fertilized embryo, decreased expression level of indicators for oxidative stress, and apoptosis in blastocysts. To our knowledge, this is the first study to demonstrate that DMEM-CM can be an optimal supplement during IVC to promote in vitro embryo development and the success rate of assisted reproduction with its anti-oxidative and anti-apoptotic effects. Abstract The quality of embryos produced by assisted reproductive techniques should be advanced by the improvement of in vitro culture conditions for successful implantation and pregnancy maintenance. We investigated the anti-oxidative effect of human adipose stem cell (ASC) conditioned medium with its optimal basal medium, Dulbecco′s modified Eagle′s medium (DMEM-CM), or keratinocyte serum-free medium (KSFM-CM) as supplements during in vitro culture (IVC) of in vitro fertilized mouse embryo. At first, preimplantation embryo development was evaluated in KSFM-CM and DMEM-CM supplemented cultures at various concentrations. The blastocyst (BL) and hatched BL formation rates were significantly increased in 5% DMEM-CM, while no difference was observed from KSFM-CM. Next, comparing the efficacy of KSFM-CM and DMEM-CM at the same concentration, DMEM-CM enhanced the developmental rate of 16 cells, morula, BL, and hatched BL. The expression level of reactive oxygen species decreased and that of glutathione increased in BL cultured with DMEM-CM, which confirms its anti-oxidative effect. Furthermore, apoptosis in BL cultured with DMEM-CM was reduced compared with that in KSFM-CM. This study demonstrated that the comparative effect of human ASC-CM made of two different basal media during mouse embryo IVC and anti-oxidative effect of 5% DMEM-CM was optimal to improve preimplantation embryo development.
Collapse
|
13
|
Bai X, Li J, Li L, Liu M, Liu Y, Cao M, Tao K, Xie S, Hu D. Extracellular Vesicles From Adipose Tissue-Derived Stem Cells Affect Notch-miR148a-3p Axis to Regulate Polarization of Macrophages and Alleviate Sepsis in Mice. Front Immunol 2020; 11:1391. [PMID: 32719678 PMCID: PMC7347748 DOI: 10.3389/fimmu.2020.01391] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 05/29/2020] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs) from adipose tissue-derived stem cells have been reported to attenuate lipopolysaccharide (LPS) induced inflammation and sepsis while the specific mechanism is unclear. This study explored the underlying molecular mechanisms of EVs from adipose tissue-derived stem cells in reducing inflammation. LPS- induced macrophage models and mice model were established to mimic inflammation in vitro and in vivo. EVs were extracted from adipose tissue-derived stem cells and identified. It was found that proinflammatory cytokines, including IL-1β, IL-6, and TNF-α, substantially decreased after EVs were applied to LPS-stimulated macrophages and mice, and thus, LPS induced M1 polarization was inhibited and sepsis was strongly alleviated. In the LPS induced macrophages, the expression of Notch signaling molecules and the activation of the NF-κB pathway were substantially decreased after the administration of EVs. Then, RBP-J -/- mice and macrophages were used. It was found that the miR-148a-3p level was significantly lower in the RBP-J -/- macrophages than in the wildtype macrophages. In the LPS induced macrophages, the increasing of miR-148a-3p was milder in the RBP-J -/- macrophages than in the wild type macrophages. Then, miR-148a-3p was overexpressed in macrophages and mice, and we found that the expression of proinflammatory cytokines was increased both in vivo and in vitro. The protective effect of EVs in LPS induced sepsis was diminished by the overexpression of miR-148a-3p. In conclusion, we proved that EVs could attenuate inflammation and further protect organ function by regulating the Notch-miR148a-3p signaling axis and then decreasing macrophage polarization to M1.
Collapse
Affiliation(s)
- Xiaozhi Bai
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Junjie Li
- Emergency Department, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lincheng Li
- Brigade 4, College of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Mingchuan Liu
- Brigade 4, College of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Yang Liu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Mengyuan Cao
- Chinese People's Liberation Army Hospital 961, Qiqihar, China
| | - Ke Tao
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Songtao Xie
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Dahai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
14
|
O Bortolazzo F, D Lucke L, de Oliveira Fujii L, Marqueti RDC, Vieira Ramos G, Theodoro V, Bombeiro AL, Felonato M, A Dalia R, D Carneiro G, Pontes Vicente C, A M Esquisatto M, A S Mendonça F, T Dos Santos GM, R Pimentel E, de Aro AA. Microcurrent and adipose-derived stem cells modulate genes expression involved in the structural recovery of transected tendon of rats. FASEB J 2020; 34:10011-10026. [PMID: 32558993 DOI: 10.1096/fj.201902942rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 05/02/2020] [Accepted: 05/05/2020] [Indexed: 01/30/2023]
Abstract
Tendon injuries are common and have a high incidence of re-rupture that can cause loss of functionality. Therapies with adipose-derived stem cells (ASC) and the microcurrent (low-intensity electrical stimulation) application present promising effects on the tissue repair. We analyzed the expression of genes and the participation of some molecules potentially involved in the structural recovery of the Achilles tendon of rats, in response to the application of both therapies, isolated and combined. The tendons were distributed in five groups: normal (N), transected (T), transected and ASC (C) or microcurrent (M) or with ASC, and microcurrent (MC). Microcurrent therapy was beneficial for tendon repair, as it was observed a statistically significant increase in the organization of the collagen fibers, with involvement of the TNC, CTGF, FN, FMDO, and COL3A1 genes as well as PCNA, IL-10, and TNF-α. ASC therapy significantly increased the TNC and FMDO genes expression with no changes in the molecular organization of collagen. With the association of therapies, a significant greater collagen fibers organization was observed with involvement of the FMOD gene. The therapies did not affect the expression of COL1A1, SMAD2, SMAD3, MKX, and EGR1 genes, nor did they influence the amount of collagen I and III, caspase-3, tenomodulin (Tnmd), and hydroxyproline. In conclusion, the application of the microcurrent isolated or associated with ASC increased the organization of the collagen fibers, which can result in a greater biomechanical resistance in relation to the tendons treated only with ASC. Future studies will be needed to demonstrate the biological effects of these therapies on the functional recovery of injured tendons.
Collapse
Affiliation(s)
- Fernanda O Bortolazzo
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas-UNICAMP, São Paulo, Brazil
| | - Letícia D Lucke
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas-UNICAMP, São Paulo, Brazil
| | - Lucas de Oliveira Fujii
- Biomedical Sciences Graduate Program, University Center of Herminio Ometto Foundation/FHO, São Paulo, Brazil
| | - Rita de Cassia Marqueti
- Graduate Program of rehabilitation science and Graduate Program of Sciences and Technology of Health and Rehabilitation Sciences, University of Brasilia (UnB), Brasília, Brazil
| | | | - Viviane Theodoro
- Biomedical Sciences Graduate Program, University Center of Herminio Ometto Foundation/FHO, São Paulo, Brazil
| | - André L Bombeiro
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas-UNICAMP, São Paulo, Brazil
| | - Maíra Felonato
- Biomedical Sciences Graduate Program, University Center of Herminio Ometto Foundation/FHO, São Paulo, Brazil
| | - Rodrigo A Dalia
- Biomedical Sciences Graduate Program, University Center of Herminio Ometto Foundation/FHO, São Paulo, Brazil
| | - Giane D Carneiro
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas-UNICAMP, São Paulo, Brazil
| | - Cristina Pontes Vicente
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas-UNICAMP, São Paulo, Brazil
| | - Marcelo A M Esquisatto
- Biomedical Sciences Graduate Program, University Center of Herminio Ometto Foundation/FHO, São Paulo, Brazil
| | - Fernanda A S Mendonça
- Biomedical Sciences Graduate Program, University Center of Herminio Ometto Foundation/FHO, São Paulo, Brazil
| | - Gláucia Maria T Dos Santos
- Biomedical Sciences Graduate Program, University Center of Herminio Ometto Foundation/FHO, São Paulo, Brazil
| | - Edson R Pimentel
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas-UNICAMP, São Paulo, Brazil
| | - Andrea A de Aro
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas-UNICAMP, São Paulo, Brazil.,Biomedical Sciences Graduate Program, University Center of Herminio Ometto Foundation/FHO, São Paulo, Brazil
| |
Collapse
|
15
|
Extensive Characterization of Mesenchymal Stem Cell Marker Expression on Freshly Isolated and In Vitro Expanded Human Adipose-Derived Stem Cells from Breast Cancer Patients. Stem Cells Int 2020; 2020:8237197. [PMID: 32655648 PMCID: PMC7320289 DOI: 10.1155/2020/8237197] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/20/2020] [Accepted: 05/27/2020] [Indexed: 02/08/2023] Open
Abstract
Variation in numbers and functions of cells in fat tissues may affect therapeutic outcomes and adverse events after autologous fat tissue grafting in postmastectomy breast cancer patients; however, the relevant information regarding cellular components is still incomplete. Phenotypic characterization of heterogeneous cell subsets in stromal vascular fraction (SVF) isolated from fat tissues by flow cytometry was also limited to a combination of few molecules. This study, therefore, developed a polychromatic staining panel for an in-depth characterization of freshly isolated SVF and expanded adipose-derived stem cells (ADSC) from the patients. ADSC were found predominant in SVF (~65% of CD45− cells) with a homogenous phenotype of CD13+CD31−CD34+CD45−CD73+CD90+CD105−CD146− (~94% of total ADSC). Endothelial progenitor cells (EPC) and pericytes were minor (~18% and ~11% of CD45− cells, respectively) with large heterogeneity. Downregulation of CD34 and upregulation of CD105 in ADSC were profound at passage 3, showing a phenotype similar to the classical mesenchymal stem cells from the bone marrow. Results from this study demonstrated that fat tissue collected from patients contains ADSC with a highly homogenous phenotype. The in vitro culture of these cells maintained their homogeneity with modified CD34 and CD105 expression, suggesting the expansion from a single population of ADSC.
Collapse
|
16
|
High Frequency of Intravenous Injection of Human Adipose Stem Cell Conditioned Medium Improved Embryo Development of Mice in Advanced Maternal Age through Antioxidant Effects. Animals (Basel) 2020; 10:ani10060978. [PMID: 32512813 PMCID: PMC7341498 DOI: 10.3390/ani10060978] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/30/2020] [Accepted: 06/03/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary In this study, we examined the anti-oxidative effect of human adipose stem cell conditioned medium (ASC-CM) on the ovary and uterus of mice in advanced maternal age (AMA) and the optimal conditions of intravenous injection for ASC-CM administration. Human ASC-CM upregulated expression of antioxidant genes, restored the quality of oocytes derived from aged ovaries and resulted in improved in vitro and in vivo embryo development. The anti-oxidative effect human ASC-CM was optimized with high frequency of administration. Comprehensively, our study successfully introduced the potential of ASC-CM as an antioxidant intervention against age-related infertility in AMA. Abstract Advanced maternal age (AMA) has become prevalent globally. With aging, weakened antioxidant defense causes loss of normal function in the ovary and uterus due to oxidative stress. Here, we aimed to improve embryo development in AMA mice by intravenous injection (IV) of human adipose stem cell conditioned medium (ASC-CM) at various frequencies and intervals as an antioxidant intervention. Four- and six-month-old female ICR (Institute of Cancer Research) mice were randomly divided into groups IV treated with human ASC-CM under different conditions, and in vitro and in vivo embryo development were evaluated. Consequently, compared to the control group, blastocyst formation rate of parthenotes was significantly promoted in 4-month-old mice and the mean number of implanted fetuses after natural mating was significantly increased by approximately two-fold in 6-month-old mice. Through gene analysis, the anti-apoptotic and anti-oxidative effects of human ASC-CMs were confirmed in the ovaries and uterus of pregnant mice at both ages. In particular, ovarian expression of gpx1 and catalase drastically increased in 6-month-old mice. Furthermore, the levels of gpx1 and catalase were further increased, with a high frequency of injection regardless of age. Thus, we demonstrated for the first time the anti-oxidative effect of human ASC-CM administration against ovarian aging and the optimal injection condition.
Collapse
|
17
|
Sarveazad A, Babahajian A, Yari A, Rayner CK, Mokhtare M, Babaei-Ghazani A, Agah S, Mahjoubi B, Shamseddin J, Yousefifard M. Combination of laser and human adipose-derived stem cells in repair of rabbit anal sphincter injury: a new therapeutic approach. Stem Cell Res Ther 2019; 10:367. [PMID: 31791407 PMCID: PMC6889595 DOI: 10.1186/s13287-019-1477-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 10/28/2019] [Accepted: 10/30/2019] [Indexed: 12/20/2022] Open
Abstract
Background Anal sphincter injury leads to fecal incontinence. Based on the regenerative capability of laser and human adipose-derived stem cells (hADSCs), this study was designed to assess the effects of co-application of these therapies on anal sphincter recovery after injury. Design Male rabbits were assigned to equal groups (n = 7) including control, sphincterotomy, sphincterotomy treated with laser (660 nm, 90 s, immediately after sphincterotomy, daily, 14 days), hADSCs (2 × 106 hADSCs injected into injured area of the sphincter immediately after sphincterotomy), and laser + hADSCs. Ninety days after sphincterotomy, manometry and electromyography were performed, sphincter collagen content was evaluated, and Ki67, myosin heavy chain (MHC), skeletal muscle alpha-actin (ACTA1), vascular endothelial growth factor A (VEGFA), and vimentin mRNA gene expression were assessed. Results The laser + hADSCs group had a higher resting pressure compared with the sphincterotomy (p < 0.0001), laser (p < 0.0001), and hADSCs (p = 0.04) groups. Maximum squeeze pressure was improved in all treated animals compared with the sphincterotomized animals (p < 0.0001), without a significant difference between treatments (p > 0.05). In the laser + hADSCs group, motor unit numbers were higher than those in the laser group (p < 0.0001) but did not differ from the hADSCs group (p = 0.075). Sphincterotomy increased collagen content, but the muscle content (p = 0.36) and collagen content (p = 0.37) were not significantly different between the laser + hADSCs and control groups. Laser + hADSCs increased ACTA1 (p = 0.001) and MHC (p < 0.0001) gene expression compared with laser or hADSCs alone and was associated with increased VEGFA (p = 0.009) and Ki67 mRNA expression (p = 0.01) and decreased vimentin mRNA expression (p < 0.0001) compared with laser. Conclusion The combination of laser and hADSCs appears more effective than either treatment alone for promoting myogenesis, angiogenesis, and functional recovery after anal sphincterotomy.
Collapse
Affiliation(s)
- Arash Sarveazad
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Asrin Babahajian
- Liver and Digestive Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Abazar Yari
- Department of Anatomy, Faculty of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Chris K Rayner
- Discipline of Medicine, University of Adelaide, Adelaide, SA, Australia.,Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA, Australia
| | - Marjan Mokhtare
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Arash Babaei-Ghazani
- Neuromusculoskeletal Research Center, Department of Physical Medicine and Rehabilitation, Iran University of Medical Sciences, Tehran, Iran
| | - Shahram Agah
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Bahar Mahjoubi
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Jebreil Shamseddin
- Infectious and Tropical Diseases Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mahmoud Yousefifard
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Scioli MG, Storti G, D'Amico F, Gentile P, Kim BS, Cervelli V, Orlandi A. Adipose-Derived Stem Cells in Cancer Progression: New Perspectives and Opportunities. Int J Mol Sci 2019; 20:3296. [PMID: 31277510 PMCID: PMC6651808 DOI: 10.3390/ijms20133296] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 12/12/2022] Open
Abstract
Growing importance has been attributed to interactions between tumors, the stromal microenvironment and adult mesenchymal stem cells. Adipose-derived stem cells (ASCs) are routinely employed in regenerative medicine and in autologous fat transfer procedures. To date, clinical trials have failed to demonstrate the potential pro-oncogenic role of ASC enrichment. Nevertheless, some pre-clinical studies from in vitro and in vivo models have suggested that ASCs act as a potential tumor promoter for different cancer cell types, and support tumor progression and invasiveness through the activation of several intracellular signals. Interaction with the tumor microenvironment and extracellular matrix remodeling, the exosomal release of pro-oncogenic factors as well as the induction of epithelial-mesenchymal transitions are the most investigated mechanisms. Moreover, ASCs have also demonstrated an elective tumor homing capacity and this tumor-targeting capacity makes them a suitable carrier for anti-cancer drug delivery. New genetic and applied nanotechnologies may help to design promising anti-cancer cell-based approaches through the release of loaded intracellular nanoparticles. These new anti-cancer therapies can more effectively target tumor cells, reaching higher local concentrations even in pharmacological sanctuaries, and thus minimizing systemic adverse drug effects. The potential interplay between ASCs and tumors and potential ASCs-based therapeutic approaches are discussed.
Collapse
Affiliation(s)
- Maria Giovanna Scioli
- Anatomic Pathology Institute, Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Gabriele Storti
- Plastic and Reconstructive Surgery, Department of Surgical Sciences, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Federico D'Amico
- Anatomic Pathology Institute, Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Pietro Gentile
- Plastic and Reconstructive Surgery, Department of Surgical Sciences, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Bong-Sung Kim
- Division of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Valerio Cervelli
- Plastic and Reconstructive Surgery, Department of Surgical Sciences, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Augusto Orlandi
- Anatomic Pathology Institute, Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy.
| |
Collapse
|
19
|
Gentile P, Garcovich S. Concise Review: Adipose-Derived Stem Cells (ASCs) and Adipocyte-Secreted Exosomal microRNA (A-SE-miR) Modulate Cancer Growth and proMote Wound Repair. J Clin Med 2019; 8:855. [PMID: 31208047 PMCID: PMC6616456 DOI: 10.3390/jcm8060855] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/05/2019] [Accepted: 06/12/2019] [Indexed: 12/13/2022] Open
Abstract
Adipose-derived stem cells (ASCs) have been routinely used from several years in regenerative surgery without any definitive statement about their potential pro-oncogenic or anti-oncogenic role. ASCs has proven to favor tumor progression in several experimental cancer models, playing a central role in regulating tumor invasiveness and metastatic potential through several mechanisms, such as the paracrine release of exosomes containing pro-oncogenic molecules and the induction of epithelial-mesenchymal transition. However, the high secretory activity and the preferential tumor-targeting make also ASCs a potentially suitable vehicle for delivery of new anti-cancer molecules in tumor microenvironment. Nanotechnologies, viral vectors, drug-loaded exosomes, and micro-RNAs (MiR) represent additional new tools that can be applied for cell-mediated drug delivery in a tumor microenvironment. Recent studies revealed that the MiR play important roles in paracrine actions on adipose-resident macrophages, and their dysregulation has been implicated in the pathogenesis of obesity, diabetes, and diabetic complications as wounds. Numerous MiR are present in adipose tissues, actively participating in the regulation of adipogenesis, adipokine secretion, inflammation, and inter-cellular communications in the local tissues. These results provide important insights into Adipocyte-secreted exosomal microRNA (A-SE-MiR) function and they suggest evaluating the potential role of A-SE-MiR in tumor progression, the mechanisms underlying ASCs-cancer cell interplay and clinical safety of ASCs-based therapies.
Collapse
Affiliation(s)
- Pietro Gentile
- Surgical Science Department, Plastic and Reconstructive Surgery Unit, University of "Tor Vergata", 00133 Rome, Italy.
| | - Simone Garcovich
- Institute of Dermatology, F. Policlinico Gemelli IRCSS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| |
Collapse
|
20
|
Fertsch S, Hagouan M, Munder B, Schulz T, Abu-Ghazaleh A, Schaberick J, Stambera P, Aldeeri M, Andree C, Thamm OC. Increased risk of recurrence associated with certain risk factors in breast cancer patients after DIEP-flap reconstruction and lipofilling-a matched cohort study with 200 patients. Gland Surg 2017; 6:315-323. [PMID: 28861370 DOI: 10.21037/gs.2017.03.11] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Lipofilling is performed in breast cancer patients to optimize the aesthetic outcome following breast reconstruction after mastectomy. Despite its common usage worldwide, little is known about the interaction of the lipoaspirate and dormant cancer cells. Up to date, no risk factors that increase the risk for cancer recurrence have been established. This study aims to identify risk factors for lipofilling candidates after breast cancer and questions the oncological safety of lipofilling in lymph node positive disease. METHODS Matched retrospective cohort study: the disease-free survival (DFS) between 100 breast cancer patients undergoing a lipofilling after their DIEP-flap reconstruction and 100 matched control patients with no subsequent lipofilling was analyzed. Further, patients were subdivided according to risk factors, which were categorized as patient-dependent factors (PDFs) and tumor-dependent factors (TDFs). DFS and hazard ratios (HR) were compared to identify potential risk factors that may increase cancer recurrence. RESULTS Median follow-up was 76.5 months from the mastectomy, and 31 months from the startpoint to the end of follow-up. Seven and eleven patients had recurrence in the lipofilling and control group, respectively, presenting with comparable DFS rates and an insignificant HR =0.57, 95% confidence interval (CI): 0.22-1.47, P=0.24. According to subgroup survival analysis, lipofilling increased the risk of recurrence in women with a positive nodal status (P=0.035) and a high-grade neoplasia (P=0.049). CONCLUSIONS No general increased recurrence risk was observed between the lipofilling and control group. The subgroup analysis identified high-grade neoplasia and positive nodal status to be a risk factor for cancer recurrence. Patients with a known node positive disease have an increased risk of occult micrometastases in their lymph nodes. Further studies are necessary to clarify whether dormant breast cancer cells in form of micrometastases in the lymph nodes can be reactivated by the factors secreted by adipose derived stem cells.
Collapse
Affiliation(s)
- Sonia Fertsch
- Department of Plastic and Reconstructive Surgery, SANA Krankenhaus Düsseldorf Gerresheim, Gräulingerstraße 120, Düsseldorf, Germany
| | - Mazen Hagouan
- Department of Plastic and Reconstructive Surgery, SANA Krankenhaus Düsseldorf Gerresheim, Gräulingerstraße 120, Düsseldorf, Germany
| | - Beatrix Munder
- Department of Plastic and Reconstructive Surgery, SANA Krankenhaus Düsseldorf Gerresheim, Gräulingerstraße 120, Düsseldorf, Germany
| | - Tino Schulz
- Department of Plastic and Reconstructive Surgery, SANA Krankenhaus Düsseldorf Gerresheim, Gräulingerstraße 120, Düsseldorf, Germany
| | - Alina Abu-Ghazaleh
- Department of Plastic and Reconstructive Surgery, SANA Krankenhaus Düsseldorf Gerresheim, Gräulingerstraße 120, Düsseldorf, Germany
| | - Julia Schaberick
- Department of Plastic and Reconstructive Surgery, SANA Krankenhaus Düsseldorf Gerresheim, Gräulingerstraße 120, Düsseldorf, Germany
| | - Peter Stambera
- Department of Plastic and Reconstructive Surgery, SANA Krankenhaus Düsseldorf Gerresheim, Gräulingerstraße 120, Düsseldorf, Germany
| | - Mohammed Aldeeri
- Department of Plastic and Reconstructive Surgery, SANA Krankenhaus Düsseldorf Gerresheim, Gräulingerstraße 120, Düsseldorf, Germany
| | - Christoph Andree
- Department of Plastic and Reconstructive Surgery, SANA Krankenhaus Düsseldorf Gerresheim, Gräulingerstraße 120, Düsseldorf, Germany
| | - Oliver Christian Thamm
- Department of Plastic and Reconstructive Surgery, SANA Krankenhaus Düsseldorf Gerresheim, Gräulingerstraße 120, Düsseldorf, Germany
| |
Collapse
|
21
|
Yeh WL, Tsai CF, Chen DR. Peri-foci adipose-derived stem cells promote chemoresistance in breast cancer. Stem Cell Res Ther 2017; 8:177. [PMID: 28750689 PMCID: PMC5532814 DOI: 10.1186/s13287-017-0630-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/20/2017] [Accepted: 07/10/2017] [Indexed: 02/07/2023] Open
Abstract
Background Mesenchymal stem cells in tumor microenvironment can influence therapeutic responses in various types of cancers. For triple negative breast cancer, chemotherapy remains the mainstay of standard treatment. Our aim was to investigate the correlation between human adipose-derived stem cells (hAdSCs) and chemoresistance in triple negative breast cancer. Method Conditioned medium was collected from hAdSCs, which was isolated from breast cancer patients who had had breast mastectomy. The expression of selected CD markers was evaluated by flow cytometry to characterize hAdSCs. By array analyses of the secreted cytokines and chemokines of hAdSCs, we identified CXCL1 that mediated doxorubicin resistance and the expression of ATP-binding cassette transporters ABCG2 in TNBC. By microRNA microarray, the association between hAdSC-mediated doxorubicin resistance in TNBC was also revealed. Results Conditioned medium collected from hAdSCs elicited doxorubicin resistance and enhanced the expression of ABCG2, which is a transporter responsible for the efflux of doxorubicin. CXCL1 secreted by hAdSCs downregulated miR-106a expression in triple negative breast cancer, and resulted in ABCG2 upregulation and doxorubicin resistance. Conclusions Our findings suggest that CXCL1 secreted by hAdSCs elicits doxorubicin resistance through miR-106a-mediated ABCG2 upregulation in triple negative breast cancer. These findings provide a better understanding of the importance of adipose-derived stem cells in breast cancer microenvironment regarding to the development of chemoresistance and reveal the potential of discovering novel therapeutic strategies to overcome drug resistance in TNBC. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0630-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wei-Lan Yeh
- Institute of New Drug Development, China Medical University, No. 91 Hsueh-Shih Road, Taichung, 40402, Taiwan.
| | - Cheng-Fang Tsai
- Department of Biotechnology, Asia University, No. 500 Lioufeng Road, Taichung, 41354, Taiwan
| | - Dar-Ren Chen
- Comprehensive Breast Cancer Center, Changhua Christian Hospital, No. 135 Nanxiao Street, Changhua, 50006, Taiwan.
| |
Collapse
|
22
|
Daquinag AC, Dadbin A, Snyder B, Wang X, Sahin AA, Ueno NT, Kolonin MG. Non-glycanated Decorin Is a Drug Target on Human Adipose Stromal Cells. MOLECULAR THERAPY-ONCOLYTICS 2017; 6:1-9. [PMID: 28607949 PMCID: PMC5458115 DOI: 10.1016/j.omto.2017.05.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 05/10/2017] [Indexed: 12/28/2022]
Abstract
Adipose stromal cells (ASCs) have been identified as a mesenchymal cell population recruited from white adipose tissue (WAT) by tumors and supporting cancer progression. We have previously reported the existence of a non-glycanated decorin isoform (ngDCN) marking mouse ASCs. We identified a peptide CSWKYWFGEC that binds to ngDCN and hence can serve as a vehicle for ASC-directed therapy delivery. We used hunter-killer peptides composed of CSWKYWFGEC and a pro-apoptotic moiety to deplete ASCs and suppress growth of mouse tumors. Here, we report the discovery of the human non-glycanated decorin isoform. We show that CSWKYWFGEC can be used as a probe to identify ASCs in human WAT and tumors. We demonstrate that human ngDCN is expressed on ASC surface. Finally, we validate ngDCN as a molecular target for pharmacological depletion of human ASCs with hunter-killer peptides. We propose that ngDCN-targeting agents could be developed for obesity and cancer treatment.
Collapse
Affiliation(s)
- Alexes C Daquinag
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Ali Dadbin
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Brad Snyder
- Department of Surgery, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Xiaoping Wang
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Section of Translational Breast Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Aysegul A Sahin
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Naoto T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Section of Translational Breast Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mikhail G Kolonin
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
23
|
Naderi N, Combellack EJ, Griffin M, Sedaghati T, Javed M, Findlay MW, Wallace CG, Mosahebi A, Butler PEM, Seifalian AM, Whitaker IS. The regenerative role of adipose-derived stem cells (ADSC) in plastic and reconstructive surgery. Int Wound J 2017; 14:112-124. [PMID: 26833722 PMCID: PMC7949873 DOI: 10.1111/iwj.12569] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 11/24/2015] [Accepted: 12/01/2015] [Indexed: 12/12/2022] Open
Abstract
The potential use of stem cell-based therapies for the repair and regeneration of various tissues and organs offers a paradigm shift in plastic and reconstructive surgery. The use of either embryonic stem cells (ESC) or induced pluripotent stem cells (iPSC) in clinical situations is limited because of regulations and ethical considerations even though these cells are theoretically highly beneficial. Adult mesenchymal stem cells appear to be an ideal stem cell population for practical regenerative medicine. Among these cells, adipose-derived stem cells (ADSC) have the potential to differentiate the mesenchymal, ectodermal and endodermal lineages and are easy to harvest. Additionally, adipose tissue yields a high number of ADSC per volume of tissue. Based on this background knowledge, the purpose of this review is to summarise and describe the proliferation and differentiation capacities of ADSC together with current preclinical data regarding the use of ADSC as regenerative tools in plastic and reconstructive surgery.
Collapse
Affiliation(s)
- Naghmeh Naderi
- Reconstructive Surgery & Regenerative Medicine Group, Institute of Life Sciences (ILS)Swansea University Medical SchoolSwanseaUK
- Welsh Centre for Burns & Plastic SurgeryABMU Health BoardSwanseaUK
| | - Emman J Combellack
- Reconstructive Surgery & Regenerative Medicine Group, Institute of Life Sciences (ILS)Swansea University Medical SchoolSwanseaUK
- Welsh Centre for Burns & Plastic SurgeryABMU Health BoardSwanseaUK
| | - Michelle Griffin
- UCL Centre for Nanotechnology and Regenerative MedicineUniversity College LondonLondonUK
| | - Tina Sedaghati
- UCL Centre for Nanotechnology and Regenerative MedicineUniversity College LondonLondonUK
| | - Muhammad Javed
- Reconstructive Surgery & Regenerative Medicine Group, Institute of Life Sciences (ILS)Swansea University Medical SchoolSwanseaUK
- Welsh Centre for Burns & Plastic SurgeryABMU Health BoardSwanseaUK
| | - Michael W Findlay
- Plastic & Reconstructive SurgeryStanford University Medical CentreStanfordCAUSA
| | | | - Afshin Mosahebi
- UCL Centre for Nanotechnology and Regenerative MedicineUniversity College LondonLondonUK
- Department of Plastic SurgeryRoyal Free NHS Foundation TrustLondonUK
| | - Peter EM Butler
- Department of Plastic SurgeryRoyal Free NHS Foundation TrustLondonUK
| | - Alexander M Seifalian
- UCL Centre for Nanotechnology and Regenerative MedicineUniversity College LondonLondonUK
| | - Iain S Whitaker
- Reconstructive Surgery & Regenerative Medicine Group, Institute of Life Sciences (ILS)Swansea University Medical SchoolSwanseaUK
- Welsh Centre for Burns & Plastic SurgeryABMU Health BoardSwanseaUK
| |
Collapse
|
24
|
Myckatyn TM, Wagner IJ, Mehrara BJ, Crosby MA, Park JE, Qaqish BF, Moore DT, Busch EL, Silva AK, Kaur S, Ollila DW, Lee CN. Cancer Risk after Fat Transfer: A Multicenter Case-Cohort Study. Plast Reconstr Surg 2017; 139:11-18. [PMID: 28027219 PMCID: PMC5428547 DOI: 10.1097/prs.0000000000002838] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Fat transfer is an increasingly popular method for refining postmastectomy breast reconstructions. However, concern persists that fat transfer may promote disease recurrence. Adipocytes are derived from adipose-derived stem cells and express adipocytokines that can facilitate active breast cancer cells in laboratory models. The authors sought to evaluate the association between fat transfer to the reconstructed breast and cancer recurrence in patients diagnosed with local or regional invasive breast cancers. METHODS A multicenter, case-cohort study was performed. Eligible patients from four centers (Memorial Sloan Kettering, M. D. Anderson Cancer Center, Alvin J. Siteman Cancer Center, and the University of Chicago) were identified by each site's institutional tumor registry or cancer data warehouse. Eligibility criteria were as follows: mastectomy with immediate breast reconstruction between 2006 and 2011, age older than 21 years, female sex, and incident diagnosis of invasive ductal carcinoma (stage I, II, or III). Cases consisted of all recurrences during the study period, and controls consisted of a 30 percent random sample of the study population. Cox proportional hazards regression was used to evaluate for association between fat transfer and time to recurrence in bivariate and multivariate models. RESULTS The time to disease recurrence unadjusted hazard ratio for fat transfer was 0.99 (95 percent CI, 0.56 to 1.7). After adjustment for age, body mass index, stage, HER2/Neu receptor status, and estrogen receptor status, the hazard ratio was 0.97 (95 percent CI, 0.54 to 1.8). CONCLUSION In this population of breast cancer patients who had mastectomy with immediate reconstruction, fat transfer was not associated with a higher risk of cancer recurrence. CLINICAL QUESTION/LEVEL OF EVIDENCE Therapeutic, III.
Collapse
Affiliation(s)
- Terence M Myckatyn
- St. Louis, Mo.; Chapel Hill, N.C.; New York, N.Y.; Houston, Texas; Chicago and Arlington Heights, Ill.; Boston, Mass.; and Columbus, Ohio
- From the Division of Plastic and Reconstructive Surgery, Washington University School of Medicine in Saint Louis, and the Alvin J. Siteman Cancer Center; the Division of Plastic and Reconstructive Surgery, the Department of Biostatistics, the Department of Epidemiology, Gillings School of Global Public Health, the Lineberger Comprehensive Cancer Center, and the Division of Surgical Oncology, Department of Surgery, University of North Carolina; the Department of Surgery, Memorial Sloan Kettering Cancer Center; the Department of Plastic Surgery, M. D. Anderson Cancer Center; the Section of Plastic and Reconstructive Surgery, University of Chicago Medicine and Biological Sciences, University of Chicago; The Plastic Surgery Foundation; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; the Department of Epidemiology, Harvard T. H. Chan School of Public Health; and Plastic and Reconstructive Surgery, Health Services Management and Policy, Ohio State University
| | - I Janelle Wagner
- St. Louis, Mo.; Chapel Hill, N.C.; New York, N.Y.; Houston, Texas; Chicago and Arlington Heights, Ill.; Boston, Mass.; and Columbus, Ohio
- From the Division of Plastic and Reconstructive Surgery, Washington University School of Medicine in Saint Louis, and the Alvin J. Siteman Cancer Center; the Division of Plastic and Reconstructive Surgery, the Department of Biostatistics, the Department of Epidemiology, Gillings School of Global Public Health, the Lineberger Comprehensive Cancer Center, and the Division of Surgical Oncology, Department of Surgery, University of North Carolina; the Department of Surgery, Memorial Sloan Kettering Cancer Center; the Department of Plastic Surgery, M. D. Anderson Cancer Center; the Section of Plastic and Reconstructive Surgery, University of Chicago Medicine and Biological Sciences, University of Chicago; The Plastic Surgery Foundation; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; the Department of Epidemiology, Harvard T. H. Chan School of Public Health; and Plastic and Reconstructive Surgery, Health Services Management and Policy, Ohio State University
| | - Babak J Mehrara
- St. Louis, Mo.; Chapel Hill, N.C.; New York, N.Y.; Houston, Texas; Chicago and Arlington Heights, Ill.; Boston, Mass.; and Columbus, Ohio
- From the Division of Plastic and Reconstructive Surgery, Washington University School of Medicine in Saint Louis, and the Alvin J. Siteman Cancer Center; the Division of Plastic and Reconstructive Surgery, the Department of Biostatistics, the Department of Epidemiology, Gillings School of Global Public Health, the Lineberger Comprehensive Cancer Center, and the Division of Surgical Oncology, Department of Surgery, University of North Carolina; the Department of Surgery, Memorial Sloan Kettering Cancer Center; the Department of Plastic Surgery, M. D. Anderson Cancer Center; the Section of Plastic and Reconstructive Surgery, University of Chicago Medicine and Biological Sciences, University of Chicago; The Plastic Surgery Foundation; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; the Department of Epidemiology, Harvard T. H. Chan School of Public Health; and Plastic and Reconstructive Surgery, Health Services Management and Policy, Ohio State University
| | - Melissa A Crosby
- St. Louis, Mo.; Chapel Hill, N.C.; New York, N.Y.; Houston, Texas; Chicago and Arlington Heights, Ill.; Boston, Mass.; and Columbus, Ohio
- From the Division of Plastic and Reconstructive Surgery, Washington University School of Medicine in Saint Louis, and the Alvin J. Siteman Cancer Center; the Division of Plastic and Reconstructive Surgery, the Department of Biostatistics, the Department of Epidemiology, Gillings School of Global Public Health, the Lineberger Comprehensive Cancer Center, and the Division of Surgical Oncology, Department of Surgery, University of North Carolina; the Department of Surgery, Memorial Sloan Kettering Cancer Center; the Department of Plastic Surgery, M. D. Anderson Cancer Center; the Section of Plastic and Reconstructive Surgery, University of Chicago Medicine and Biological Sciences, University of Chicago; The Plastic Surgery Foundation; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; the Department of Epidemiology, Harvard T. H. Chan School of Public Health; and Plastic and Reconstructive Surgery, Health Services Management and Policy, Ohio State University
| | - Julie E Park
- St. Louis, Mo.; Chapel Hill, N.C.; New York, N.Y.; Houston, Texas; Chicago and Arlington Heights, Ill.; Boston, Mass.; and Columbus, Ohio
- From the Division of Plastic and Reconstructive Surgery, Washington University School of Medicine in Saint Louis, and the Alvin J. Siteman Cancer Center; the Division of Plastic and Reconstructive Surgery, the Department of Biostatistics, the Department of Epidemiology, Gillings School of Global Public Health, the Lineberger Comprehensive Cancer Center, and the Division of Surgical Oncology, Department of Surgery, University of North Carolina; the Department of Surgery, Memorial Sloan Kettering Cancer Center; the Department of Plastic Surgery, M. D. Anderson Cancer Center; the Section of Plastic and Reconstructive Surgery, University of Chicago Medicine and Biological Sciences, University of Chicago; The Plastic Surgery Foundation; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; the Department of Epidemiology, Harvard T. H. Chan School of Public Health; and Plastic and Reconstructive Surgery, Health Services Management and Policy, Ohio State University
| | - Bahjat F Qaqish
- St. Louis, Mo.; Chapel Hill, N.C.; New York, N.Y.; Houston, Texas; Chicago and Arlington Heights, Ill.; Boston, Mass.; and Columbus, Ohio
- From the Division of Plastic and Reconstructive Surgery, Washington University School of Medicine in Saint Louis, and the Alvin J. Siteman Cancer Center; the Division of Plastic and Reconstructive Surgery, the Department of Biostatistics, the Department of Epidemiology, Gillings School of Global Public Health, the Lineberger Comprehensive Cancer Center, and the Division of Surgical Oncology, Department of Surgery, University of North Carolina; the Department of Surgery, Memorial Sloan Kettering Cancer Center; the Department of Plastic Surgery, M. D. Anderson Cancer Center; the Section of Plastic and Reconstructive Surgery, University of Chicago Medicine and Biological Sciences, University of Chicago; The Plastic Surgery Foundation; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; the Department of Epidemiology, Harvard T. H. Chan School of Public Health; and Plastic and Reconstructive Surgery, Health Services Management and Policy, Ohio State University
| | - Dominic T Moore
- St. Louis, Mo.; Chapel Hill, N.C.; New York, N.Y.; Houston, Texas; Chicago and Arlington Heights, Ill.; Boston, Mass.; and Columbus, Ohio
- From the Division of Plastic and Reconstructive Surgery, Washington University School of Medicine in Saint Louis, and the Alvin J. Siteman Cancer Center; the Division of Plastic and Reconstructive Surgery, the Department of Biostatistics, the Department of Epidemiology, Gillings School of Global Public Health, the Lineberger Comprehensive Cancer Center, and the Division of Surgical Oncology, Department of Surgery, University of North Carolina; the Department of Surgery, Memorial Sloan Kettering Cancer Center; the Department of Plastic Surgery, M. D. Anderson Cancer Center; the Section of Plastic and Reconstructive Surgery, University of Chicago Medicine and Biological Sciences, University of Chicago; The Plastic Surgery Foundation; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; the Department of Epidemiology, Harvard T. H. Chan School of Public Health; and Plastic and Reconstructive Surgery, Health Services Management and Policy, Ohio State University
| | - Evan L Busch
- St. Louis, Mo.; Chapel Hill, N.C.; New York, N.Y.; Houston, Texas; Chicago and Arlington Heights, Ill.; Boston, Mass.; and Columbus, Ohio
- From the Division of Plastic and Reconstructive Surgery, Washington University School of Medicine in Saint Louis, and the Alvin J. Siteman Cancer Center; the Division of Plastic and Reconstructive Surgery, the Department of Biostatistics, the Department of Epidemiology, Gillings School of Global Public Health, the Lineberger Comprehensive Cancer Center, and the Division of Surgical Oncology, Department of Surgery, University of North Carolina; the Department of Surgery, Memorial Sloan Kettering Cancer Center; the Department of Plastic Surgery, M. D. Anderson Cancer Center; the Section of Plastic and Reconstructive Surgery, University of Chicago Medicine and Biological Sciences, University of Chicago; The Plastic Surgery Foundation; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; the Department of Epidemiology, Harvard T. H. Chan School of Public Health; and Plastic and Reconstructive Surgery, Health Services Management and Policy, Ohio State University
| | - Amanda K Silva
- St. Louis, Mo.; Chapel Hill, N.C.; New York, N.Y.; Houston, Texas; Chicago and Arlington Heights, Ill.; Boston, Mass.; and Columbus, Ohio
- From the Division of Plastic and Reconstructive Surgery, Washington University School of Medicine in Saint Louis, and the Alvin J. Siteman Cancer Center; the Division of Plastic and Reconstructive Surgery, the Department of Biostatistics, the Department of Epidemiology, Gillings School of Global Public Health, the Lineberger Comprehensive Cancer Center, and the Division of Surgical Oncology, Department of Surgery, University of North Carolina; the Department of Surgery, Memorial Sloan Kettering Cancer Center; the Department of Plastic Surgery, M. D. Anderson Cancer Center; the Section of Plastic and Reconstructive Surgery, University of Chicago Medicine and Biological Sciences, University of Chicago; The Plastic Surgery Foundation; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; the Department of Epidemiology, Harvard T. H. Chan School of Public Health; and Plastic and Reconstructive Surgery, Health Services Management and Policy, Ohio State University
| | - Surinder Kaur
- St. Louis, Mo.; Chapel Hill, N.C.; New York, N.Y.; Houston, Texas; Chicago and Arlington Heights, Ill.; Boston, Mass.; and Columbus, Ohio
- From the Division of Plastic and Reconstructive Surgery, Washington University School of Medicine in Saint Louis, and the Alvin J. Siteman Cancer Center; the Division of Plastic and Reconstructive Surgery, the Department of Biostatistics, the Department of Epidemiology, Gillings School of Global Public Health, the Lineberger Comprehensive Cancer Center, and the Division of Surgical Oncology, Department of Surgery, University of North Carolina; the Department of Surgery, Memorial Sloan Kettering Cancer Center; the Department of Plastic Surgery, M. D. Anderson Cancer Center; the Section of Plastic and Reconstructive Surgery, University of Chicago Medicine and Biological Sciences, University of Chicago; The Plastic Surgery Foundation; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; the Department of Epidemiology, Harvard T. H. Chan School of Public Health; and Plastic and Reconstructive Surgery, Health Services Management and Policy, Ohio State University
| | - David W Ollila
- St. Louis, Mo.; Chapel Hill, N.C.; New York, N.Y.; Houston, Texas; Chicago and Arlington Heights, Ill.; Boston, Mass.; and Columbus, Ohio
- From the Division of Plastic and Reconstructive Surgery, Washington University School of Medicine in Saint Louis, and the Alvin J. Siteman Cancer Center; the Division of Plastic and Reconstructive Surgery, the Department of Biostatistics, the Department of Epidemiology, Gillings School of Global Public Health, the Lineberger Comprehensive Cancer Center, and the Division of Surgical Oncology, Department of Surgery, University of North Carolina; the Department of Surgery, Memorial Sloan Kettering Cancer Center; the Department of Plastic Surgery, M. D. Anderson Cancer Center; the Section of Plastic and Reconstructive Surgery, University of Chicago Medicine and Biological Sciences, University of Chicago; The Plastic Surgery Foundation; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; the Department of Epidemiology, Harvard T. H. Chan School of Public Health; and Plastic and Reconstructive Surgery, Health Services Management and Policy, Ohio State University
| | - Clara N Lee
- St. Louis, Mo.; Chapel Hill, N.C.; New York, N.Y.; Houston, Texas; Chicago and Arlington Heights, Ill.; Boston, Mass.; and Columbus, Ohio
- From the Division of Plastic and Reconstructive Surgery, Washington University School of Medicine in Saint Louis, and the Alvin J. Siteman Cancer Center; the Division of Plastic and Reconstructive Surgery, the Department of Biostatistics, the Department of Epidemiology, Gillings School of Global Public Health, the Lineberger Comprehensive Cancer Center, and the Division of Surgical Oncology, Department of Surgery, University of North Carolina; the Department of Surgery, Memorial Sloan Kettering Cancer Center; the Department of Plastic Surgery, M. D. Anderson Cancer Center; the Section of Plastic and Reconstructive Surgery, University of Chicago Medicine and Biological Sciences, University of Chicago; The Plastic Surgery Foundation; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; the Department of Epidemiology, Harvard T. H. Chan School of Public Health; and Plastic and Reconstructive Surgery, Health Services Management and Policy, Ohio State University
| |
Collapse
|
25
|
Arshad Z, Karmen L, Choudhary R, Smith JA, Branford OA, Brindley DA, Pettitt D, Davies BM. Cell assisted lipotransfer in breast augmentation and reconstruction: A systematic review of safety, efficacy, use of patient reported outcomes and study quality. JPRAS Open 2016; 10:5-20. [PMID: 28066801 PMCID: PMC5193151 DOI: 10.1016/j.jpra.2016.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 08/14/2016] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Cell assisted lipotransfer serves as a novel technique for both breast reconstruction and breast augmentation. This systematic review assesses the efficacy, safety and use of patient reported outcome measures in studies involving cell assisted lipotransfer. We also carry out an objective assessment of study quality focussing on recruitment, follow-up and provide an up-to-date clinical trial landscaping analysis. METHODS Key electronic databases were searched according to PRISMA guidelines and pre-defined inclusion and exclusion criteria. Two independent reviewers examined the retrieved publications and performed data extraction. RESULTS 3980 publications were identified. Following screening, 11 studies were included for full review, representing a total of 336 patients with a follow-up time ranging from six to 42 months. A degree of variation was noted in graft retention and reported satisfaction levels, although there were only three comparative studies with conflicting results. Complications occurred at a rate of 37%. Additionally, there was a paucity of objective outcomes assessments (e.g. 3D assessment modalities or validated patient reported outcome measures) in the selected studies. CONCLUSIONS Cell assisted lipotransfer is a surgical technique that is currently employed sparingly within the plastic & reconstructive surgery community. Presently, further technical and outcome standardization is required, in addition to rigorous randomized controlled trials and supporting long-term follow-up data to better determine procedural safety and efficacy. Routine use of more objective outcome measures, particularly 3D assessments and validated patient reported outcome measures, will also help facilitate wider clinical adoption and establish procedural utility.
Collapse
Affiliation(s)
- Zeeshaan Arshad
- School of Medicine, University of St. Andrews, St. Andrews, North Haugh, UK
- The Oxford – UCL Centre for the Advancement of Sustainable Medical Innovation (CASMI), The University of Oxford, Oxford, UK
| | - Lindsey Karmen
- School of Medicine, University of St. Andrews, St. Andrews, North Haugh, UK
| | | | - James A. Smith
- The Oxford – UCL Centre for the Advancement of Sustainable Medical Innovation (CASMI), The University of Oxford, Oxford, UK
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Windmill Road, Oxford, UK
| | | | - David A. Brindley
- The Oxford – UCL Centre for the Advancement of Sustainable Medical Innovation (CASMI), The University of Oxford, Oxford, UK
- Said Business School, University of Oxford, Park End Street, Oxford, UK
- Centre for Behavioral Medicine, UCL School of Pharmacy, University College London, Brunswick Square, London, UK
- Harvard Stem Cell Institute, Divinity Avenue, Cambridge, MA, USA
- USCF-Stanford Center of Excellence in Regulatory Science and Innovation (CERSI), 4th Street, USA
- Department of Pediatrics, University of Oxford, Headley Way, UK
| | - David Pettitt
- The Oxford – UCL Centre for the Advancement of Sustainable Medical Innovation (CASMI), The University of Oxford, Oxford, UK
- Department of Pediatrics, University of Oxford, Headley Way, UK
| | - Benjamin M. Davies
- The Oxford – UCL Centre for the Advancement of Sustainable Medical Innovation (CASMI), The University of Oxford, Oxford, UK
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Windmill Road, Oxford, UK
| |
Collapse
|
26
|
Zhang Y, Kolonin MG. Cytokine signaling regulating adipose stromal cell trafficking. Adipocyte 2016; 5:369-374. [PMID: 27994950 DOI: 10.1080/21623945.2016.1220452] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 07/27/2016] [Accepted: 07/28/2016] [Indexed: 01/31/2023] Open
Abstract
Adipocyte progenitors, known as adipose stromal cells (ASC), can become mobilized, recruited by tumors, and contribute to cancer progression. Mechanisms underlying ASC trafficking have remained obscure. We recently reported that CXCL1 expressed by cancer cells chemoattracts ASC expressing CXCR1 in obesity. As a candidate mechanism of CXCL1 activation, we identified interleukin (IL)-22, systemic circulation of which is increased in obesity. It has been reported that IL-22 signaling through IL-22R is upstream of CXCL1. Here, we provide evidence that IL-22 expression by leukocytes infiltrating WAT and IL-22R expression by tumors is obesity-dependent. We propose that obesity-associated adipocyte death and the resulting recruitment of leukocytes triggers the IL-22 signaling cascade that induces CXCL1 secretion by cancer cells responsible for ASC trafficking to tumors.
Collapse
|
27
|
Frese L, Dijkman PE, Hoerstrup SP. Adipose Tissue-Derived Stem Cells in Regenerative Medicine. Transfus Med Hemother 2016; 43:268-274. [PMID: 27721702 DOI: 10.1159/000448180] [Citation(s) in RCA: 278] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 07/04/2016] [Indexed: 12/15/2022] Open
Abstract
In regenerative medicine, adult stem cells are the most promising cell types for cell-based therapies. As a new source for multipotent stem cells, human adipose tissue has been introduced. These so called adipose tissue-derived stem cells (ADSCs) are considered to be ideal for application in regenerative therapies. Their main advantage over mesenchymal stem cells derived from other sources, e.g. from bone marrow, is that they can be easily and repeatable harvested using minimally invasive techniques with low morbidity. ADSCs are multipotent and can differentiate into various cell types of the tri-germ lineages, including e.g. osteocytes, adipocytes, neural cells, vascular endothelial cells, cardiomyocytes, pancreatic β-cells, and hepatocytes. Interestingly, ADSCs are characterized by immunosuppressive properties and low immunogenicity. Their secretion of trophic factors enforces the therapeutic and regenerative outcome in a wide range of applications. Taken together, these particular attributes of ADSCs make them highly relevant for clinical applications. Consequently, the therapeutic potential of ADSCs is enormous. Therefore, this review will provide a brief overview of the possible therapeutic applications of ADSCs with regard to their differentiation potential into the tri-germ lineages. Moreover, the relevant advancements made in the field, regulatory aspects as well as other challenges and obstacles will be highlighted.
Collapse
Affiliation(s)
- Laura Frese
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Petra E Dijkman
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Simon P Hoerstrup
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland; Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands; Wyss Translational Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
28
|
Seo BR, Bhardwaj P, Choi S, Gonzalez J, Andresen Eguiluz RC, Wang K, Mohanan S, Morris PG, Du B, Zhou XK, Vahdat LT, Verma A, Elemento O, Hudis CA, Williams RM, Gourdon D, Dannenberg AJ, Fischbach C. Obesity-dependent changes in interstitial ECM mechanics promote breast tumorigenesis. Sci Transl Med 2016; 7:301ra130. [PMID: 26290412 DOI: 10.1126/scitranslmed.3010467] [Citation(s) in RCA: 245] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Obesity and extracellular matrix (ECM) density are considered independent risk and prognostic factors for breast cancer. Whether they are functionally linked is uncertain. We investigated the hypothesis that obesity enhances local myofibroblast content in mammary adipose tissue and that these stromal changes increase malignant potential by enhancing interstitial ECM stiffness. Indeed, mammary fat of both diet- and genetically induced mouse models of obesity were enriched for myofibroblasts and stiffness-promoting ECM components. These differences were related to varied adipose stromal cell (ASC) characteristics because ASCs isolated from obese mice contained more myofibroblasts and deposited denser and stiffer ECMs relative to ASCs from lean control mice. Accordingly, decellularized matrices from obese ASCs stimulated mechanosignaling and thereby the malignant potential of breast cancer cells. Finally, the clinical relevance and translational potential of our findings were supported by analysis of patient specimens and the observation that caloric restriction in a mouse model reduces myofibroblast content in mammary fat. Collectively, these findings suggest that obesity-induced interstitial fibrosis promotes breast tumorigenesis by altering mammary ECM mechanics with important potential implications for anticancer therapies.
Collapse
Affiliation(s)
- Bo Ri Seo
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Priya Bhardwaj
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Siyoung Choi
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Jacqueline Gonzalez
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | | | - Karin Wang
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA. Department of Materials Science and Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Sunish Mohanan
- Department of Biological and Biomedical Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Patrick G Morris
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Baoheng Du
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Xi K Zhou
- Department of Healthcare Policy and Research, Weill Cornell Medical College, New York, NY 10065, USA
| | - Linda T Vahdat
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Akanksha Verma
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Olivier Elemento
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Clifford A Hudis
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Rebecca M Williams
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Delphine Gourdon
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Andrew J Dannenberg
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Claudia Fischbach
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA. Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
29
|
Aronowitz JA, Lockhart RA, Hakakian CS. Mechanical versus enzymatic isolation of stromal vascular fraction cells from adipose tissue. SPRINGERPLUS 2015; 4:713. [PMID: 26636001 PMCID: PMC4656256 DOI: 10.1186/s40064-015-1509-2] [Citation(s) in RCA: 162] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 11/04/2015] [Indexed: 02/07/2023]
Abstract
Clinical use of adipose-derived stem cells (ASCs) for a variety of indications is rapidly expanding in medicine. Most commonly, ASCs are isolated at the point of care from lipoaspirate tissue as the stromal vascular fraction (SVF). The cells are immediately administered to the patient as an injection or used to enrich fat grafts. Isolation of ASCs from adipose tissue is a relatively simple process performed routinely in cell biology laboratories, but isolation at the point of care for immediate clinical administration requires special methodology to prevent contamination, ensure integrity of clinical research and comply with regulatory requirements. A lack of practical laboratory experience, regulatory uncertainty and a relative paucity of objective published data can make selection of the optimum separation method for specific indications a difficult task for the clinician and can discourage clinical adoption. In this paper, we discuss the processes which can be used to separate SVF cells from fat tissue. We compare the various mechanical and enzymatic methods. We discuss the practical considerations involved in selecting an appropriate method from a clinical perspective. Studies consistently show that breakdown of the extracellular matrix achieved with proteolytic enzymes affords significantly greater efficiency to the separation process. SVF isolated through mechanical methods is equally safe, less costly and less time consuming but the product contains a higher frequency of blood mononuclear cells and fewer progenitor cells. Mechanical methods can provide a low cost, rapid and simple alternative to enzymatic isolation methods, and are attractive when smaller quantities of ASCs are sufficient.
Collapse
Affiliation(s)
- Joel A. Aronowitz
- />Cedars-Sinai Medical Center, Los Angeles, USA
- />University Stem Cell Center, 8635 W 3rd St. Suite 1090W, Los Angeles, CA 90048 USA
- />USC, Keck School of Medicine, Los Angeles, USA
| | - Ryan A. Lockhart
- />University Stem Cell Center, 8635 W 3rd St. Suite 1090W, Los Angeles, CA 90048 USA
| | - Cloe S. Hakakian
- />University Stem Cell Center, 8635 W 3rd St. Suite 1090W, Los Angeles, CA 90048 USA
| |
Collapse
|
30
|
Ma L, Wen H, Jian X, Liao H, Sui Y, Liu Y, Xu G. Cell-assisted lipotransfer in the clinical treatment of facial soft tissue deformity. Plast Surg (Oakv) 2015; 23:199-202. [PMID: 26361629 DOI: 10.4172/plastic-surgery.1000926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Cosmetic surgeons have experimented with a variety of substances to improve soft tissue deformities of the face. Autologous fat grafting provides significant advantages over other modalities because it leaves no scar, is easy to use and is well tolerated by most patients. Autologous fat grafting has become one of the most popular techniques in the field of facial plastic surgery. Unfortunately, there are still two major problems affecting survival rate and development: revascularization after transplantion; and cell reservation proliferation and survival. Since Zuk and Yosra developed a technology based on adipose-derived stem cells and cell-assisted lipotrophy, researchers have hoped that this technology would promote the survival and reduce the absorption of grafted fat cells. Autologous adipose-derived stem cells may have great potential in skin repair applications, aged skin rejuvenation and other aging-related skin lesion treatments. Recently, the study of adipose-derived stem cells has gained increased attention. More researchers have started to adopt this technology in the clinical treatment of facial soft tissue deformity. The present article reviews the history of facial soft tissue augmentation and the advent of adipose-derived stem cells in the area of the clinical treatment of facial soft tissue deformity.
Collapse
Affiliation(s)
- Li Ma
- Department of Plastic Surgery, First Affiliated Hospital of Nanchang University
| | - Huicai Wen
- Department of Plastic Surgery, First Affiliated Hospital of Nanchang University
| | | | | | - Yunpeng Sui
- Nanchang University, Nanchang, Jiangxi, China
| | - Yanping Liu
- Nanchang University, Nanchang, Jiangxi, China
| | - Guizhen Xu
- Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
31
|
The role of adipose-derived stem cells in breast cancer progression and metastasis. Stem Cells Int 2015; 2015:120949. [PMID: 26000019 PMCID: PMC4427098 DOI: 10.1155/2015/120949] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 12/26/2014] [Indexed: 12/15/2022] Open
Abstract
Conventional breast cancer extirpation involves resection of parts of or the whole gland, resulting in asymmetry and disfiguration. Given the unsatisfactory aesthetic outcomes, patients often desire postmastectomy reconstructive procedures. Autologous fat grafting has been proposed for reconstructive purposes for decades to restore form and anatomy after mastectomy. Fat has the inherent advantage of being autologous tissue and the most natural-appearing filler, but given its inconsistent engraftment and retention rates, it lacks reliability. Implementation of autologous fat grafts with cellular adjuncts, such as multipotent adipose-derived stem cells (ADSCs), has shown promising results. However, it is pertinent and critical to question whether these cells could promote any residual tumor cells to proliferate, differentiate, or metastasize or even induce de novo carcinogenesis. Thus far, preclinical and clinical study findings are discordant. A trend towards potential promotion of both breast cancer growth and invasion by ADSCs found in basic science studies was indeed not confirmed in clinical trials. Whether experimental findings eventually correlate with or will be predictive of clinical outcomes remains unclear. Herein, we aimed to concisely review current experimental findings on the interaction of mesenchymal stem cells and breast cancer, mainly focusing on ADSCs as a promising tool for regenerative medicine, and discuss the implications in clinical translation.
Collapse
|
32
|
Stem cells from adipose tissue and breast cancer: hype, risks and hope. Br J Cancer 2015; 112:419-23. [PMID: 25584493 PMCID: PMC4453662 DOI: 10.1038/bjc.2014.657] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 10/21/2014] [Accepted: 12/03/2014] [Indexed: 12/20/2022] Open
Abstract
Several recent papers have generated new hope about the use of white adipose tissue (WAT)-derived progenitor cells for soft tissue reconstruction in a variety of diseases including breast cancer (BC), a procedure that is increasingly used worldwide. We revised the available literature about WAT cells and BC. In the BC field, we believe that the hype for the exciting results in terms of WAT progenitor cell engraftment and tissue augmentation should be tempered when considering the recent and abundant preclinical studies, indicating that WAT progenitors may promote BC growth and metastasis. White adipose tissue progenitors can contribute to tumour vessels, pericytes and adipocytes, and were found to stimulate local and metastatic BC progression in several murine models. Moreover, there are clinical retrospective data showing a significant increase in the local recurrence frequency in patients with intraepithelial neoplasia who received a lipofilling procedure for breast reconstruction compared with controls. Retrospective and prospective clinical trials are warranted to investigate in depth the safety of this procedure in BC. Preclinical models should be used to find mechanisms able to inhibit the tumour-promoting activity of WAT progenitors while sparing their tissue reconstruction potential.
Collapse
|
33
|
Lohsiriwat V, Petit J. Nipple Sparing Mastectomy: from prophylactic to therapeutic standard. Gland Surg 2014; 1:75-9. [PMID: 25083428 DOI: 10.3978/j.issn.2227-684x.2012.06.02] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Accepted: 06/20/2012] [Indexed: 01/24/2023]
Affiliation(s)
- Visnu Lohsiriwat
- 1 Department of Plastic and Reconstructive Surgery, European Institute of Oncology, Milan, Italy ; 2 Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University Bangkok, Thailand
| | - JeanYves Petit
- 1 Department of Plastic and Reconstructive Surgery, European Institute of Oncology, Milan, Italy ; 2 Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University Bangkok, Thailand
| |
Collapse
|
34
|
Tsuji W, Rubin JP, Marra KG. Adipose-derived stem cells: Implications in tissue regeneration. World J Stem Cells 2014; 6:312-321. [PMID: 25126381 PMCID: PMC4131273 DOI: 10.4252/wjsc.v6.i3.312] [Citation(s) in RCA: 266] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 05/16/2014] [Accepted: 06/11/2014] [Indexed: 02/06/2023] Open
Abstract
Adipose-derived stem cells (ASCs) are mesenchymal stem cells (MSCs) that are obtained from abundant adipose tissue, adherent on plastic culture flasks, can be expanded in vitro, and have the capacity to differentiate into multiple cell lineages. Unlike bone marrow-derived MSCs, ASCs can be obtained from abundant adipose tissue by a minimally invasive procedure, which results in a high number of cells. Therefore, ASCs are promising for regenerating tissues and organs damaged by injury and diseases. This article reviews the implications of ASCs in tissue regeneration.
Collapse
|
35
|
Bielli A, Scioli MG, Gentile P, Agostinelli S, Tarquini C, Cervelli V, Orlandi A. Adult adipose-derived stem cells and breast cancer: a controversial relationship. SPRINGERPLUS 2014; 3:345. [PMID: 25089245 PMCID: PMC4117859 DOI: 10.1186/2193-1801-3-345] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 06/11/2014] [Indexed: 01/13/2023]
Abstract
Breast cancer is the most common cancer in women and autologous fat grafting is an important clinical application in treatment of post-surgical deformities. The simplicity of fat grafting procedures and the absence of subsequent visible scar prompted an increasing interest for this technique. The plasticity of adipose-derived stem cells (ASCs) obtained from stromal vascular fraction (SVF) of adult adipose tissue provided exciting perspectives for regenerative medicine and surgery. The recent discovery that SVF/ASC enrichment further ameliorates clinical efficacy of grafting ASCs suggest as ASC-mediated new adipogenesis and vasculogenesis. ASC adipogenic differentiation involves Akt activity and EGFRs, FGFRs, ERbB2 receptor-mediated pathways that also play a pivotal role in the regulation of breast cancer growth. Moreover, the finding that platelet-derived growth factors and hormones improved long-term maintenance of fat grafting raises new concerns for their use during breast reconstruction after cancer surgery. However, it remains unclear whether grafted or resident ASCs may increase the risk of de novo cancer development or recurrence. Preliminary follow-up studies seem to support the efficacy and safety of SVF/ASCs enrichment and the additional benefit from the combined use of autologous platelet-derived growth factors and hormones during breast reconstruction procedures. In the present review we highlighted the complex interplay between resident or grafted ASCs, mature adipocytes, dormant or active breast cancer cells and tumor microenvironment. Actually, data concerning the permissive role of ASCs on breast cancer progression are contrasting, although no clear evidence speaking against their use exists.
Collapse
Affiliation(s)
- Alessandra Bielli
- />Anatomic Pathology, Tor Vergata University of Rome, Via Montpellier, 00133 Rome, Italy
| | - Maria Giovanna Scioli
- />Anatomic Pathology, Tor Vergata University of Rome, Via Montpellier, 00133 Rome, Italy
| | - Pietro Gentile
- />Plastic Surgery, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Sara Agostinelli
- />Anatomic Pathology, Tor Vergata University of Rome, Via Montpellier, 00133 Rome, Italy
| | - Chiara Tarquini
- />Anatomic Pathology, Tor Vergata University of Rome, Via Montpellier, 00133 Rome, Italy
| | - Valerio Cervelli
- />Plastic Surgery, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Augusto Orlandi
- />Anatomic Pathology, Tor Vergata University of Rome, Via Montpellier, 00133 Rome, Italy
| |
Collapse
|
36
|
Bertolini F. Adipose tissue and breast cancer progression: a link between metabolism and cancer. Breast 2014; 22 Suppl 2:S48-9. [PMID: 24074792 DOI: 10.1016/j.breast.2013.07.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION/AIMS Obesity, an excess accumulation of adipose tissue occurring in mammalians when caloric intake exceeds energy expenditure, is associated with an increased incidence, morbidity and mortality from several types of neoplastic diseases including postmenopausal breast cancer. METHODS AND RESULTS Several investigators have recently studied the role of human white adipose tissue (WAT) progenitors in preclinical models of breast cancer. WAT progenitors were found to promote breast cancer local growth, angiogenesis, EMT, migration and metastatic spreading. Breast cancer patients with intraepithelial neoplasia who received autologous WAT cells for breast reconstruction after surgical removal of breast cancer showed an increased risk of recurrence of local events when compared to controls. DISCUSSION/CONCLUSION There is an urgent need for a better understanding of the role of WAT progenitors in breast cancer local and metastatic growth. A rigorous cancer screening and follow-up of patients enrolled for WAT progenitor-based therapies should be implemented.
Collapse
Affiliation(s)
- Francesco Bertolini
- Laboratory of Hematology-Oncology, European Institute of Oncology, Via Ripamonti 435, 20141 Milan, Italy.
| |
Collapse
|
37
|
Rowan BG, Gimble JM, Sheng M, Anbalagan M, Jones RK, Frazier TP, Asher M, Lacayo EA, Friedlander PL, Kutner R, Chiu ES. Human adipose tissue-derived stromal/stem cells promote migration and early metastasis of triple negative breast cancer xenografts. PLoS One 2014; 9:e89595. [PMID: 24586900 PMCID: PMC3938488 DOI: 10.1371/journal.pone.0089595] [Citation(s) in RCA: 144] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 01/22/2014] [Indexed: 01/01/2023] Open
Abstract
Background Fat grafting is used to restore breast defects after surgical resection of breast tumors. Supplementing fat grafts with adipose tissue-derived stromal/stem cells (ASCs) is proposed to improve the regenerative/restorative ability of the graft and retention. However, long term safety for ASC grafting in proximity of residual breast cancer cells is unknown. The objective of this study was to determine the impact of human ASCs derived from abdominal lipoaspirates of three donors, on a human breast cancer model that exhibits early metastasis. Methodology/Principal Findings Human MDA-MB-231 breast cancer cells represents “triple negative” breast cancer that exhibits early micrometastasis to multiple mouse organs [1]. Human ASCs were derived from abdominal adipose tissue from three healthy female donors. Indirect co-culture of MDA-MB-231 cells with ASCs, as well as direct co-culture demonstrated that ASCs had no effect on MDA-MB-231 growth. Indirect co-culture, and ASC conditioned medium (CM) stimulated migration of MDA-MB-231 cells. ASC/RFP cells from two donors co-injected with MDA-MB-231/GFP cells exhibited a donor effect for stimulation of primary tumor xenografts. Both ASC donors stimulated metastasis. ASC/RFP cells were viable, and integrated with MDA-MB-231/GFP cells in the tumor. Tumors from the co-injection group of one ASC donor exhibited elevated vimentin, matrix metalloproteinase-9 (MMP-9), IL-8, VEGF and microvessel density. The co-injection group exhibited visible metastases to the lung/liver and enlarged spleen not evident in mice injected with MDA-MB-231/GFP alone. Quantitation of the total area of GFP fluorescence and human chromosome 17 DNA in mouse organs, H&E stained paraffin sections and fluorescent microscopy confirmed multi-focal metastases to lung/liver/spleen in the co-injection group without evidence of ASC/RFP cells. Conclusions Human ASCs derived from abdominal lipoaspirates of two donors stimulated metastasis of MDA-MB-231 breast tumor xenografts to multiple mouse organs. MDA-MB-231 tumors co-injected with ASCs from one donor exhibited partial EMT, expression of MMP-9, and increased angiogenesis.
Collapse
Affiliation(s)
- Brian G. Rowan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
- * E-mail: (BGR); (ESC)
| | - Jeffrey M. Gimble
- Stem Cell Biology Laboratory, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Mei Sheng
- Department of Otolaryngology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Muralidharan Anbalagan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Ryan K. Jones
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Trivia P. Frazier
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Majdouline Asher
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Eduardo A. Lacayo
- Department of Otolaryngology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Paul L. Friedlander
- Department of Otolaryngology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Robert Kutner
- Gene Therapy Program, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Ernest S. Chiu
- Department of Plastic Surgery, New York University Langone Medical Center, New York, New York, United States of America
- * E-mail: (BGR); (ESC)
| |
Collapse
|
38
|
Hoppe DL, Ueberreiter K, Surlemont Y, Peltoniemi H, Stabile M, Kauhanen S. Breast reconstruction de novo by water-jet assisted autologous fat grafting--a retrospective study. GERMAN MEDICAL SCIENCE : GMS E-JOURNAL 2013; 11:Doc17. [PMID: 24403878 PMCID: PMC3884560 DOI: 10.3205/000185] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 11/15/2013] [Indexed: 12/20/2022]
Abstract
BACKGROUND Autologous fat grafting has become a frequent, simple, reproducible and low-risk technique for revisional or partial breast reconstruction. The presented European multicenter study describes an optimized treatment and follow-up protocol for the de novo breast reconstruction after total mastectomy by lipotransfer alone. METHODS A retrospective European multicenter trial included 135 procedures on 28 (35 breasts) postmastectomy patients (mean 52.4 years). All women were treated with the water-jet assisted fat grafting method (BEAULI™) combined with additional procedures (NAC reconstruction, contralateral mastoplasty) and evaluated with at least 6 months follow-up (mean 2.6 years). Sonography or mammography, clinical examination, patient questionnaire (10-point Likert scale) and digital photographs were carried out. RESULTS On average the patients received 4 to 6 procedures each with a single volume of 159 ml (±61 ml) over 21 months (range 9 months to 2.5 years). In total 1,020 ml (±515 ml) fat were grafted till a complete breast reconstruction was achieved. Irradiated patients needed a significantly higher volume than non-irradiated (p<0.041). Main treatment complications were liponecrosis (2.59%), infection (0.74%) and granuloma (0.74%). Patient satisfaction was overall high to very high (96%) and confirmed the good aesthetic results (68%) and the natural softness, contour and shape of the reconstructed breast. CONCLUSIONS A complete breast reconstruction with large volume fat grafting is alternatively possible to standard techniques in selected cases. It takes at least 4 to 6 lipotransfers in the course of 2 years. Patients with prior radiotherapy may require even up to 8 sessions over nearly 3 years of treatment.
Collapse
Affiliation(s)
- Delia Letizia Hoppe
- BG Unfallklinik Tübingen, Klinik für Hand-, Plastische, Rekonstruktive und Verbrennungschirurgie, Tübingen, Germany
| | | | - Yves Surlemont
- Clinique Saint Antoine, Rouen University Hospital, Paediatric and Dermatologic Plastic Surgery, Bois Guillaume, France
| | | | | | - Susanna Kauhanen
- Helsinki University Hospital, Department of Plastic and Reconstructive Surgery, Helsinki, Finland
| |
Collapse
|
39
|
Goldhirsch A, Winer EP, Coates AS, Gelber RD, Piccart-Gebhart M, Thürlimann B, Senn HJ. Personalizing the treatment of women with early breast cancer: highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann Oncol 2013; 24:2206-23. [PMID: 23917950 PMCID: PMC3755334 DOI: 10.1093/annonc/mdt303] [Citation(s) in RCA: 2649] [Impact Index Per Article: 220.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 07/01/2013] [Accepted: 07/02/2013] [Indexed: 02/07/2023] Open
Abstract
The 13th St Gallen International Breast Cancer Conference (2013) Expert Panel reviewed and endorsed substantial new evidence on aspects of the local and regional therapies for early breast cancer, supporting less extensive surgery to the axilla and shorter durations of radiation therapy. It refined its earlier approach to the classification and management of luminal disease in the absence of amplification or overexpression of the Human Epidermal growth factor Receptor 2 (HER2) oncogene, while retaining essentially unchanged recommendations for the systemic adjuvant therapy of HER2-positive and 'triple-negative' disease. The Panel again accepted that conventional clinico-pathological factors provided a surrogate subtype classification, while noting that in those areas of the world where multi-gene molecular assays are readily available many clinicians prefer to base chemotherapy decisions for patients with luminal disease on these genomic results rather than the surrogate subtype definitions. Several multi-gene molecular assays were recognized as providing accurate and reproducible prognostic information, and in some cases prediction of response to chemotherapy. Cost and availability preclude their application in many environments at the present time. Broad treatment recommendations are presented. Such recommendations do not imply that each Panel member agrees: indeed, among more than 100 questions, only one (trastuzumab duration) commanded 100% agreement. The various recommendations in fact carried differing degrees of support, as reflected in the nuanced wording of the text below and in the votes recorded in supplementary Appendix S1, available at Annals of Oncology online. Detailed decisions on treatment will as always involve clinical consideration of disease extent, host factors, patient preferences and social and economic constraints.
Collapse
Affiliation(s)
- A Goldhirsch
- International Breast Cancer Study Group, Division of Medical Oncology, European Institute of Oncology, Milan, Italy.
| | | | | | | | | | | | | |
Collapse
|
40
|
Orecchioni S, Gregato G, Martin-Padura I, Reggiani F, Braidotti P, Mancuso P, Calleri A, Quarna J, Marighetti P, Aldeni C, Pruneri G, Martella S, Manconi A, Petit JY, Rietjens M, Bertolini F. Complementary populations of human adipose CD34+ progenitor cells promote growth, angiogenesis, and metastasis of breast cancer. Cancer Res 2013; 73:5880-91. [PMID: 23918796 DOI: 10.1158/0008-5472.can-13-0821] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Obesity is associated with an increased frequency, morbidity, and mortality of several types of neoplastic diseases, including postmenopausal breast cancer. We found that human adipose tissue contains two populations of progenitors with cooperative roles in breast cancer. CD45(-)CD34(+)CD31(+)CD13(-)CCRL2(+) endothelial cells can generate mature endothelial cells and capillaries. Their cancer-promoting effect in the breast was limited in the absence of CD45(-)CD34(+)CD31(-)CD13(+)CD140b(+) mesenchymal progenitors/adipose stromal cells (ASC), which generated pericytes and were more efficient than endothelial cells in promoting local tumor growth. Both endothelial cells and ASCs induced epithelial-to-mesenchymal transition (EMT) gene expression in luminal breast cancer cells. Endothelial cells (but not ASCs) migrated to lymph nodes and to contralateral nascent breast cancer lesions where they generated new vessels. In vitro and in vivo, endothelial cells were more efficient than ASCs in promoting tumor migration and in inducing metastases. Granulocyte colony-stimulating factor (G-CSF) effectively mobilized endothelial cells (but not ASCs), and the addition of chemotherapy and/or of CXCR4 inhibitors did not increase endothelial cell or ASC blood mobilization. Our findings suggest that adipose tissue progenitor cells cooperate in driving progression and metastatic spread of breast cancer.
Collapse
Affiliation(s)
- Stefania Orecchioni
- Authors' Affiliations: Laboratory of Hematology-Oncology, Department of Pathology, and Division of Plastic Surgery, European Institute of Oncology; and Division of Pathology, San Paolo University Hospital, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|