264 results on '"Bruno Péault"'
Search Results
2. Umbilical cord artery-derived perivascular stem cells for treatment of ovarian failure through CD146 signaling
- Author
-
Lu Xu, Yanjun Yang, Lingling Zhang, Guijun Yan, Shiyuan Li, Yifan Li, Yali Hu, Lijun Ding, Bruno Péault, and Haixiang Sun
- Subjects
Medicine ,Biology (General) ,QH301-705.5 - Published
- 2022
- Full Text
- View/download PDF
3. Aging modulates the effects of ischemic injury upon mesenchymal cells within the renal interstitium and microvasculature
- Author
-
Isaac W. Shaw, Eoin D. O'Sullivan, Angela O. Pisco, Gary Borthwick, Kevin M. Gallagher, Bruno Péault, Jeremy Hughes, and David A. Ferenbach
- Subjects
aging ,fibrosis ,ischemia ,kidney ,mesenchyme ,pericyte ,Medicine (General) ,R5-920 ,Cytology ,QH573-671 - Abstract
Abstract The renal mesenchyme contains heterogeneous cells, including interstitial fibroblasts and pericytes, with key roles in wound healing. Although healing is impaired in aged kidneys, the effect of age and injury on the mesenchyme remains poorly understood. We characterized renal mesenchymal cell heterogeneity in young vs old animals and after ischemia‐reperfusion‐injury (IRI) using multiplex immunolabeling and single cell transcriptomics. Expression patterns of perivascular cell markers (α‐SMA, CD146, NG2, PDGFR‐α, and PDGFR‐β) correlated with their interstitial location. PDGFR‐α and PDGFR‐β co‐expression labeled renal myofibroblasts more efficiently than the current standard marker α‐SMA, and CD146 was a superior murine renal pericyte marker. Three renal mesenchymal subtypes; pericytes, fibroblasts, and myofibroblasts, were recapitulated with data from two independently performed single cell transcriptomic analyzes of murine kidneys, the first dataset an aging cohort and the second dataset injured kidneys following IRI. Mesenchymal cells segregated into subtypes with distinct patterns of expression with aging and following injury. Baseline uninjured old kidneys resembled post‐ischemic young kidneys, with this phenotype further exaggerated following IRI. These studies demonstrate that age modulates renal perivascular/interstitial cell marker expression and transcriptome at baseline and in response to injury and provide tools for the histological and transcriptomic analysis of renal mesenchymal cells, paving the way for more accurate classification of renal mesenchymal cell heterogeneity and identification of age‐specific pathways and targets.
- Published
- 2021
- Full Text
- View/download PDF
4. Transplantation of human endometrial perivascular cells with elevated CYR61 expression induces angiogenesis and promotes repair of a full-thickness uterine injury in rat
- Author
-
Zhongxun Li, Guijun Yan, Qiang Diao, Fei Yu, Xin’an Li, Xiaoqiang Sheng, Yong Liu, Yimin Dai, Huaijun Zhou, Xin Zhen, Yali Hu, Bruno Péault, Lijun Ding, Haixiang Sun, and Hairong Li
- Subjects
Endometrial perivascular cells ,CYR61 ,Uterine injury ,Neovascularization ,Fertility ,Medicine (General) ,R5-920 ,Biochemistry ,QD415-436 - Abstract
Abstract Background Disruptions of angiogenesis can have a significant effect on the healing of uterine scars. Human endometrial perivascular cells (CD146+PDGFRβ+) function as stem cells in the endometrium. Cysteine-rich angiogenic inducer 61 (CYR61) plays an important role in vascular development. The purpose of this study was to observe the effects of the transplantation of human endometrial perivascular cells (En-PSCs) overexpressing CYR61 on structural and functional regeneration in rat models of partial full-thickness uterine excision. Methods We first sorted human En-PSCs from endometrial single-cell suspensions by flow cytometry. Human En-PSCs expressing low or high levels of CYR61 were then generated via transfection with a CYR61-specific small interfering ribonucleic acid (si-CYR61) construct or overexpression plasmid. To establish a rat model of uterine injury, a subset of uterine wall was then resected from each uterine horn in experimental animals. Female rats were randomly assigned to five groups, including a sham-operated group and four repair groups that received either PBS loaded on a collagen scaffold (collagen/PBS), En-PSCs loaded on a collagen scaffold (collagen/En-PSCs), En-PSCs with low CYR61 expression loaded on a collagen scaffold (collagen/si-CYR61 En-PSCs), and En-PSCs overexpressing CYR61 loaded on a collagen scaffold (collagen/ov-CYR61 En-PSCs). These indicated constructs were sutured in the injured uterine area to replace the excised segment. On days 30 and 90 after transplantation, a subset of rats in each group was sacrificed, and uterine tissue was recovered and serially sectioned. Hematoxylin and eosin staining and immunohistochemical staining were then performed. Finally, the remaining rats of each group were mated with fertile male rats on day 90 for a 2-week period. Results Sorted En-PSCs expressed all recognized markers of mesenchymal stem cells (MSCs), including CD10, CD13, CD44, CD73, CD90, and CD105, and exhibited differentiation potential toward adipocytes, osteoblasts, and neuron-like cells. Compared with En-PSCs and En-PSCs with low CYR61 expression, En-PSCs with elevated CYR61 expression enhanced angiogenesis by in vitro co-culture assays. At day 90 after transplantation, blood vessel density in the collagen/ov-CYR61 En-PSCs group (11.667 ± 1.287) was greater than that in the collagen/En-PSCs group (7.167 ± 0.672) (P
- Published
- 2019
- Full Text
- View/download PDF
5. Lysosomal protein surface expression discriminates fat- from bone-forming human mesenchymal precursor cells
- Author
-
Jiajia Xu, Yiyun Wang, Ching-Yun Hsu, Stefano Negri, Robert J Tower, Yongxing Gao, Ye Tian, Takashi Sono, Carolyn A Meyers, Winters R Hardy, Leslie Chang, Shuaishuai Hu, Nusrat Kahn, Kristen Broderick, Bruno Péault, and Aaron W James
- Subjects
perivascular stem cell ,mesenchymal stem cell ,osteogenesis ,adipogenesis ,CD107a/LAMP1 ,exocytosis ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Tissue resident mesenchymal stem/stromal cells (MSCs) occupy perivascular spaces. Profiling human adipose perivascular mesenchyme with antibody arrays identified 16 novel surface antigens, including endolysosomal protein CD107a. Surface CD107a expression segregates MSCs into functionally distinct subsets. In culture, CD107alow cells demonstrate high colony formation, osteoprogenitor cell frequency, and osteogenic potential. Conversely, CD107ahigh cells include almost exclusively adipocyte progenitor cells. Accordingly, human CD107alow cells drove dramatic bone formation after intramuscular transplantation in mice, and induced spine fusion in rats, whereas CD107ahigh cells did not. CD107a protein trafficking to the cell surface is associated with exocytosis during early adipogenic differentiation. RNA sequencing also suggested that CD107alow cells are precursors of CD107ahigh cells. These results document the molecular and functional diversity of perivascular regenerative cells, and show that relocation to cell surface of a lysosomal protein marks the transition from osteo- to adipogenic potential in native human MSCs, a population of substantial therapeutic interest.
- Published
- 2020
- Full Text
- View/download PDF
6. Five Decades Later, Are Mesenchymal Stem Cells Still Relevant?
- Author
-
Mario Gomez-Salazar, Zaniah N. Gonzalez-Galofre, Joan Casamitjana, Mihaela Crisan, Aaron W. James, and Bruno Péault
- Subjects
tissue engineering ,mesenchymal stem cell ,pericyte ,cell therapy ,adventitia ,Biotechnology ,TP248.13-248.65 - Abstract
Mesenchymal stem cells are culture-derived mesodermal progenitors isolatable from all vascularized tissues. In spite of multiple fundamental, pre-clinical and clinical studies, the native identity and role in tissue repair of MSCs have long remained elusive, with MSC selection in vitro from total cell suspensions essentially unchanged as a mere primary culture for half a century. Recent investigations have helped understand the tissue origin of these progenitor cells, and uncover alternative effects of MSCs on tissue healing via growth factor secretion and interaction with the immune system. In this review, we describe current trends in MSC biology and discuss how these may improve the use of these therapeutic cells in tissue engineering and regenerative medicine.
- Published
- 2020
- Full Text
- View/download PDF
7. Transcriptionally and Functionally Distinct Mesenchymal Subpopulations Are Generated from Human Pluripotent Stem Cells
- Author
-
Chee Jia Chin, Suwen Li, Mirko Corselli, David Casero, Yuhua Zhu, Chong Bin He, Reef Hardy, Bruno Péault, and Gay M. Crooks
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Summary: Various mesenchymal cell types have been identified as critical components of the hematopoietic stem/progenitor cell (HSPC) niche. Although several groups have described the generation of mesenchyme from human pluripotent stem cells (hPSCs), the capacity of such cells to support hematopoiesis has not been reported. Here, we demonstrate that distinct mesenchymal subpopulations co-emerge from mesoderm during hPSC differentiation. Despite co-expression of common mesenchymal markers (CD73, CD105, CD90, and PDGFRβ), a subset of cells defined as CD146hiCD73hi expressed genes associated with the HSPC niche and supported the maintenance of functional HSPCs ex vivo, while CD146loCD73lo cells supported differentiation. Stromal support of HSPCs was contact dependent and mediated in part through high JAG1 expression and low WNT signaling. Molecular profiling revealed significant transcriptional similarity between hPSC-derived CD146++ and primary human CD146++ perivascular cells. The derivation of functionally diverse types of mesenchyme from hPSCs opens potential avenues to model the HSPC niche and develop PSC-based therapies. : Crooks and colleagues demonstrated a previously underappreciated functional and molecular heterogeneity in mesenchyme generated from human pluripotent stem cells. Two mesenchymal subsets were distinguished by the reciprocal expression of CD146, CD73, and CD140a. CD146hiCD73hi mesenchyme supported self-renewing hematopoietic stem and progenitor cells (HSPCs), expressed markers of the HSPC niche, and shared a similar molecular signature with primary human adult pericytes. Keywords: pluripotent stem cell, mesenchyme, hematopoietic stem cell niche, pericyte biology, directed differentiation, mesoderm
- Published
- 2018
- Full Text
- View/download PDF
8. Improving cartilage phenotype from differentiated pericytes in tunable peptide hydrogels
- Author
-
Enateri V. Alakpa, Vineetha Jayawarna, Karl E. V. Burgess, Christopher C. West, Bruno Péault, Rein V. Ulijn, and Matthew J. Dalby
- Subjects
Medicine ,Science - Abstract
Abstract Differentiation of stem cells to chondrocytes in vitro usually results in a heterogeneous phenotype. This is evident in the often detected over expression of type X collagen which, in hyaline cartilage structure is not characteristic of the mid-zone but of the deep-zone ossifying tissue. Methods to better match cartilage developed in vitro to characteristic in vivo features are therefore highly desirable in regenerative medicine. This study compares phenotype characteristics between pericytes, obtained from human adipose tissue, differentiated using diphenylalanine/serine (F2/S) peptide hydrogels with the more widely used chemical induced method for chondrogenesis. Significantly higher levels of type II collagen were noted when pericytes undergo chondrogenesis in the hydrogel in the absence of induction media. There is also a balanced expression of collagen relative to aggrecan production, a feature which was biased toward collagen production when cells were cultured with induction media. Lastly, metabolic profiles of each system show considerable overlap between both differentiation methods but subtle differences which potentially give rise to their resultant phenotype can be ascertained. The study highlights how material and chemical alterations in the cellular microenvironment have wide ranging effects on resultant tissue type.
- Published
- 2017
- Full Text
- View/download PDF
9. Isolation and characterization of equine native MSC populations
- Author
-
Cristina L. Esteves, Tara A. Sheldrake, Simone P. Mesquita, Juan J. Pesántez, Timothy Menghini, Lucy Dawson, Bruno Péault, and F. Xavier Donadeu
- Subjects
Pericyte ,Equine ,Horse ,Adventitial cells ,CD146 ,CD34 ,Medicine (General) ,R5-920 ,Biochemistry ,QD415-436 - Abstract
Abstract Background In contrast to humans in which mesenchymal stem/stromal cell (MSC) therapies are still largely in the clinical trial phase, MSCs have been used therapeutically in horses for over 15 years, thus constituting a valuable preclinical model for humans. In human tissues, MSCs have been shown to originate from perivascular cells, namely pericytes and adventitial cells, which are identified by the presence of the cell surface markers CD146 and CD34, respectively. In contrast, the origin of MSCs in equine tissues has not been established, preventing the isolation and culture of defined cell populations in that species. Moreover, a comparison between perivascular CD146+ and CD34+ cell populations has not been performed in any species. Methods Immunohistochemistry was used to identify adventitial cells (CD34+) and pericytes (CD146+) and to determine their localization in relation to MSCs in equine tissues. Isolation of CD34+ (CD34+/CD146–/CD144–/CD45–) and CD146+ (CD146+/CD34–/CD144–/CD45–) cell fractions from equine adipose tissue was achieved by fluorescence-activated cell sorting. The isolated cell fractions were cultured and analyzed for the expression of MSC markers, using qPCR and flow cytometry, and for the ability to undergo trilineage differentiation. Angiogenic properties were analyzed in vivo using a chorioallantoic membrane (CAM) assay. Results Both CD34+ and CD146+ cells displayed typical MSC features, namely growth in uncoated tissue culture dishes, clonal growth when seeded at low density, expression of typical MSC markers, and multipotency shown by the capacity for trilineage differentiation. Of note, CD146+ cells were distinctly angiogenic compared with CD34+ and non-sorted cells (conventional MSCs), demonstrated by the induction of blood vessels in a CAM assay, expression of elevated levels of VEGFA and ANGPT1, and association with vascular networks in cocultures with endothelial cells, indicating that CD146+ cells maintain a pericyte phenotype in culture. Conclusion This study reports for the first time the successful isolation and culture of CD146+ and CD34+ cell populations from equine tissues. Characterization of these cells evidenced their distinct properties and MSC-like phenotype, and identified CD146+ cells as distinctly angiogenic, which may provide a novel source for enhanced regenerative therapies.
- Published
- 2017
- Full Text
- View/download PDF
10. Skeletal Myogenesis in vitro
- Author
-
William Chen and Bruno Péault
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Mature skeletal myofibers are elongated and multinucleated cells. Many stem/progenitor cell types, including committed muscle stem (satellite cells) and progenitor (myoblasts) cells, muscle-derived stem cells, myogenic endothelial cells, and mesenchymal stem/stromal cells, have been shown to exhibit skeletal myogenesis under appropriate inductive conditions. Committed muscle stem/progenitor cells and multipotent stem/progenitor cells which have skeletal myogenic capacity can typically be differentiated into skeletal myofibers in vitro following extended low-serum exposure. Differentiated cells exhibit distinct fiber-like elongated morphology with multiple nuclei and express unique muscle molecular markers indicating myogenesis, including desmin (early) and fast- and/or slow-myosin heavy chain (mature).
- Published
- 2015
- Full Text
- View/download PDF
11. Regenerative Translation of Human Blood-Vessel-Derived MSC Precursors
- Author
-
William C. W. Chen, Bruno Péault, and Johnny Huard
- Subjects
Internal medicine ,RC31-1245 - Abstract
Mesenchymal stem/stromal cells (MSCs) represent a promising adult progenitor cell source for tissue repair and regeneration. Their mysterious identity in situ has gradually been unveiled by the accumulating evidence indicating an association between adult multipotent stem/progenitor cells and vascular/perivascular niches. Using immunohistochemistry and fluorescence-activated cell sorting, we and other groups have prospectively identified and purified subpopulations of multipotent precursor cells associated with the blood vessels within multiple human organs. The three precursor subsets, myogenic endothelial cells (MECs), pericytes (PCs), and adventitial cells (ACs), are located, respectively, in the three structural tiers of typical blood vessels: intima, media, and adventitia. MECs, PCs, and ACs have been extensively characterized in prior studies and are currently under investigation for their therapeutic potentials in preclinical animal models. In this review, we will briefly discuss the identification, isolation, and characterization of these human blood-vessel-derived stem cells (hBVSCs) and summarize the current status of regenerative applications of hBVSC subsets.
- Published
- 2015
- Full Text
- View/download PDF
12. Cellular Kinetics of Perivascular MSC Precursors
- Author
-
William C. W. Chen, Tea Soon Park, Iain R. Murray, Ludovic Zimmerlin, Lorenza Lazzari, Johnny Huard, and Bruno Péault
- Subjects
Internal medicine ,RC31-1245 - Abstract
Mesenchymal stem/stromal cells (MSCs) and MSC-like multipotent stem/progenitor cells have been widely investigated for regenerative medicine and deemed promising in clinical applications. In order to further improve MSC-based stem cell therapeutics, it is important to understand the cellular kinetics and functional roles of MSCs in the dynamic regenerative processes. However, due to the heterogeneous nature of typical MSC cultures, their native identity and anatomical localization in the body have remained unclear, making it difficult to decipher the existence of distinct cell subsets within the MSC entity. Recent studies have shown that several blood-vessel-derived precursor cell populations, purified by flow cytometry from multiple human organs, give rise to bona fide MSCs, suggesting that the vasculature serves as a systemic reservoir of MSC-like stem/progenitor cells. Using individually purified MSC-like precursor cell subsets, we and other researchers have been able to investigate the differential phenotypes and regenerative capacities of these contributing cellular constituents in the MSC pool. In this review, we will discuss the identification and characterization of perivascular MSC precursors, including pericytes and adventitial cells, and focus on their cellular kinetics: cell adhesion, migration, engraftment, homing, and intercellular cross-talk during tissue repair and regeneration.
- Published
- 2013
- Full Text
- View/download PDF
13. Isolation of Myogenic Stem Cells from Cultures of Cryopreserved Human Skeletal Muscle
- Author
-
Bo Zheng, Chien-Wen Chen, Guangheng Li, Seth D. Thompson, Minakshi Poddar, Bruno Péault, and Johnny Huard Ph.D.
- Subjects
Medicine - Abstract
We demonstrate that subpopulations of adult human skeletal muscle-derived stem cells, myogenic endothelial cells (MECs), and perivascular stem cells (PSCs) can be simultaneously purified by fluorescence-activated cell sorting (FACS) from cryopreserved human primary skeletal muscle cell cultures (cryo-hPSMCs). For FACS isolation, we utilized a combination of cell lineage markers: the myogenic cell marker CD56, the endothelial cell marker UEA-1 receptor (UEA-1R), and the perivascular cell marker CD146. MECs expressing all three cell lineage markers (CD56 + UEA-1R + CD146 + /CD45 - ) and PSCs expressing only CD146 (CD146 + /CD45 - CD56 - UEA-1R - ) were isolated by FACS. To evaluate their myogenic capacities, the sorted cells, with and without expansion in culture, were transplanted into the cardiotoxin-injured skeletal muscles of immunodeficient mice. The purified MECs exhibited the highest regenerative capacity in the injured mouse muscles among all cell fractions tested, while PSCs remained superior to myoblasts and the unpurified primary skeletal muscle cells. Our findings show that both MECs and PSCs retain their high myogenic potentials after in vitro expansion, cryopreservation, and FACS sorting. The current study demonstrates that myogenic stem cells are prospectively isolatable from long-term cryopreserved primary skeletal muscle cell cultures. We emphasize the potential application of this new approach to extract therapeutic stem cells from human muscle cells cryogenically banked for clinical purposes.
- Published
- 2012
- Full Text
- View/download PDF
14. Venous graft-derived cells participate in peripheral nerve regeneration.
- Author
-
Mitra Lavasani, Sebastian Gehrmann, Burhan Gharaibeh, Katherine A Clark, Robert A Kaufmann, Bruno Péault, Robert J Goitz, and Johnny Huard
- Subjects
Medicine ,Science - Abstract
Based on growing evidence that some adult multipotent cells necessary for tissue regeneration reside in the walls of blood vessels and the clinical success of vein wrapping for functional repair of nerve damage, we hypothesized that the repair of nerves via vein wrapping is mediated by cells migrating from the implanted venous grafts into the nerve bundle.To test the hypothesis, severed femoral nerves of rats were grafted with venous grafts from animals of the opposite sex. Nerve regeneration was impaired when decellularized or irradiated venous grafts were used in comparison to untreated grafts, supporting the involvement of venous graft-derived cells in peripheral nerve repair. Donor cells bearing Y chromosomes integrated into the area of the host injured nerve and participated in remyelination and nerve regeneration. The regenerated nerve exhibited proper axonal myelination, and expressed neuronal and glial cell markers.These novel findings identify the mechanism by which vein wrapping promotes nerve regeneration.
- Published
- 2011
- Full Text
- View/download PDF
15. Bioengineered niches that recreate physiological extracellular matrix organisation to support long-term haematopoietic stem cells
- Author
-
Hannah Donnelly, Ewan Ross, Yinbo Xiao, Rio Hermantara, Aqeel F. Taqi, W. Sebastian Doherty-Boyd, Jennifer Cassels, Penelope. M. Tsimbouri, Karen M. Dunn, Jodie Hay, Annie Cheng, R. M. Dominic Meek, Nikhil Jain, Christopher West, Helen Wheadon, Alison M. Michie, Bruno Peault, Adam G. West, Manuel Salmeron-Sanchez, and Matthew J. Dalby
- Subjects
Science - Abstract
Abstract Long-term reconstituting haematopoietic stem cells (LT-HSCs) are used to treat blood disorders via stem cell transplantation. The very low abundance of LT-HSCs and their rapid differentiation during in vitro culture hinders their clinical utility. Previous developments using stromal feeder layers, defined media cocktails, and bioengineering have enabled HSC expansion in culture, but of mostly short-term HSCs and progenitor populations at the expense of naive LT-HSCs. Here, we report the creation of a bioengineered LT-HSC maintenance niche that recreates physiological extracellular matrix organisation, using soft collagen type-I hydrogels to drive nestin expression in perivascular stromal cells (PerSCs). We demonstrate that nestin, which is expressed by HSC-supportive bone marrow stromal cells, is cytoprotective and, via regulation of metabolism, is important for HIF-1α expression in PerSCs. When CD34+ve HSCs were added to the bioengineered niches comprising nestin/HIF-1α expressing PerSCs, LT-HSC numbers were maintained with normal clonal and in vivo reconstitution potential, without media supplementation. We provide proof-of-concept that our bioengineered niches can support the survival of CRISPR edited HSCs. Successful editing of LT-HSCs ex vivo can have potential impact on the treatment of blood disorders.
- Published
- 2024
- Full Text
- View/download PDF
16. Functional Heterogeneity of Perivascular Precursor Cells
- Author
-
Bruno Péault, Stefano Negri, Jiajia Xu, Aaron W. James, Ching Yun Hsu, Yiyun Wang, and Bradley Presson
- Subjects
medicine.anatomical_structure ,Cellular heterogeneity ,Precursor cell ,Mesenchymal stem cell ,Tunica Adventitia ,medicine ,Adipose tissue ,General Medicine ,Biology ,Stem cell ,Perivascular space ,Stem cell niche ,Cell biology - Abstract
Tissue-resident mesenchymal precursor cells occupy perivascular spaces. Two main perivascular locations have been described for forerunners of cultured mesenchymal stem cells, including a subendothelial location (termed pericytes) and cells within the tunica adventitia (termed adventitial cells). Emerging data have made clear the cellular heterogeneity within the perivascular stem cell niche. This brief review focuses on two issues. First, the inter-tissue heterogeneity of pericytes, including the theoretical consideration of pericytes as tissue-specific stem cells. Second, the emerging intra-tissue heterogeneity of adventitial cells (a.k.a adventicytes). New data suggest that these fibroblast-like cells within the fat tissue are functionally diverse. Markers that segregate for functionally relevant subsets of adventitial cells will be reviewed.
- Published
- 2020
17. Human perivascular stem cells prevent bone graft resorption in osteoporotic contexts by inhibiting osteoclast formation
- Author
-
Stefano Negri, Kristen P. Broderick, Ginny Ching Yun Hsu, Aaron W. James, Yiyun Wang, Jiajia Xu, Qizhi Qin, Takashi Sono, Seungyong Lee, Bruno Péault, Carolyn A. Meyers, and Kenneth W. Witwer
- Subjects
musculoskeletal diseases ,0301 basic medicine ,Stromal cell ,Nude ,bone graft ,Osteoclasts ,Bone resorption ,Rats, Nude ,03 medical and health sciences ,Paracrine signalling ,0302 clinical medicine ,Osteoclast ,Tissue Engineering and Regenerative Medicine ,pericyte ,perivascular stem cell ,medicine ,Animals ,Humans ,Bone Resorption ,Bone regeneration ,mesenchymal stem cell ,spine fusion ,Chemistry ,Stem Cells ,Mesenchymal stem cell ,adipose stem cell ,Osteoblast ,Cell Biology ,General Medicine ,Rats ,Cell biology ,Resorption ,030104 developmental biology ,medicine.anatomical_structure ,osteoclast ,Osteoporosis ,Female ,Transcriptome ,030217 neurology & neurosurgery ,Stem Cell Transplantation ,Developmental Biology - Abstract
The vascular wall stores mesenchymal progenitor cells which are able to induce bone regeneration, via direct and paracrine mechanisms. Although much is known regarding perivascular cell regulation of osteoblasts, their regulation of osteoclasts, and by extension utility in states of high bone resorption, is not known. Here, human perivascular stem cells (PSCs) were used as a means to prevent autograft resorption in a gonadectomy‐induced osteoporotic spine fusion model. Furthermore, the paracrine regulation by PSCs of osteoclast formation was evaluated, using coculture, conditioned medium, and purified extracellular vesicles. Results showed that PSCs when mixed with autograft bone induce an increase in osteoblast:osteoclast ratio, promote bone matrix formation, and prevent bone graft resorption. The confluence of these factors resulted in high rates of fusion in an ovariectomized rat lumbar spine fusion model. Application of PSCs was superior across metrics to either the use of unpurified, culture‐defined adipose‐derived stromal cells or autograft bone alone. Under coculture conditions, PSCs negatively regulated osteoclast formation and did so via secreted, nonvesicular paracrine factors. Total RNA sequencing identified secreted factors overexpressed by PSCs which may explain their negative regulation of graft resorption. In summary, PSCs reduce osteoclast formation and prevent bone graft resorption in high turnover states such as gonadectomy‐induced osteoporosis., Perivascular stem cells (PSCs) derived from adipose tissue prevent bone graft resorption and enhance arthrodesis in a preclinical osteoporotic spinal fusion model. PSCs inhibit osteoclasts formation via paracrine secretion of negative regulators in a high bone turnover environment.
- Published
- 2020
18. Skeletogenic Capacity of Human Perivascular Stem Cells Obtained Via Magnetic-Activated Cell Sorting
- Author
-
Greg Asatrian, Erin Zou, Aaron W. James, Leslie Chang, Leititia Zhang, Catherine Ding, Bruno Péault, Min Lee, Noah Yan, Kristen P. Broderick, Jiajia Xu, Yiyun Wang, and Carolyn A. Meyers
- Subjects
Adult ,Stromal cell ,medicine.medical_treatment ,0206 medical engineering ,Population ,Biomedical Engineering ,Antigens, CD34 ,Bioengineering ,Cell Separation ,02 engineering and technology ,Biology ,Biochemistry ,Biomaterials ,03 medical and health sciences ,Osteogenesis ,medicine ,Humans ,Cell Lineage ,education ,030304 developmental biology ,Wound Healing ,0303 health sciences ,education.field_of_study ,Magnetic-activated cell sorting ,Magnetic Phenomena ,Stem Cells ,Skull ,Mesenchymal stem cell ,Cell Differentiation ,Original Articles ,Stem-cell therapy ,Cell sorting ,020601 biomedical engineering ,Cell biology ,Adipose Tissue ,CD146 ,Stem cell ,Biomarkers - Abstract
Human perivascular stem/stromal cells (PSC) are a multipotent mesenchymal progenitor cell population defined by their perivascular residence. PSC are increasingly studied for their application in skeletal regenerative medicine. PSC from subcutaneous white adipose tissue are most commonly isolated via fluorescence-activated cell sorting (FACS), and defined as a bipartite population of CD146(+)CD34(−)CD31(−)CD45(−) pericytes and CD34(+)CD146(−)CD31(−)CD45(−) adventitial cells. FACS poses several challenges for clinical translation, including requirements for facilities, equipment, and personnel. The purpose of this study is to identify if magnetic-activated cell sorting (MACS) is a feasible method to derive PSC, and to determine if MACS-derived PSC are comparable to our previous experience with FACS-derived PSC. In brief, CD146(+) pericytes and CD34(+) adventitial cells were enriched from human lipoaspirate using a multistep column approach. Next, cell identity and purity were analyzed by flow cytometry. In vitro multilineage differentiation studies were performed with MACS-defined PSC subsets. Finally, in vivo application was performed in nonhealing calvarial bone defects in Scid mice. Results showed that human CD146(+) pericytes and CD34(+) adventitial cells may be enriched by MACS, with defined purity, anticipated cell surface marker expression, and capacity for multilineage differentiation. In vivo, MACS-derived PSC induce ossification of bone defects. These data document the feasibility of a MACS approach for the enrichment and application of PSC in the field of tissue engineering and regenerative medicine. IMPACT STATEMENT: Our findings suggest that perivascular stem/stromal cells, and in particular adventitial cells, may be isolated by magnetic-activated cell sorting and applied as an uncultured autologous stem cell therapy in a same-day setting for bone defect repair.
- Published
- 2019
19. PDGFRα marks distinct perivascular populations with different osteogenic potential within adipose tissue
- Author
-
Ye Tian, Aaron W. James, Yiyun Wang, Jiajia Xu, Carolyn A. Meyers, Kristen P. Broderick, Yongxing Gao, and Bruno Péault
- Subjects
Male ,0301 basic medicine ,Receptor, Platelet-Derived Growth Factor alpha ,Stromal cell ,CD34 ,Adipose tissue ,Biology ,Bone morphogenetic protein 2 ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Osteogenesis ,medicine ,Animals ,Humans ,Progenitor cell ,Mesenchymal stem cell ,Cell Differentiation ,Osteoblast ,Cell Biology ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Adipose Tissue ,Molecular Medicine ,Pericyte ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
The perivascular niche within adipose tissue is known to house multipotent cells, including osteoblast precursors. However, the identity of perivascular subpopulations that may mineralize or ossify most readily is not known. Here, we utilize inducible PDGFRα (platelet-derived growth factor alpha) reporter animals to identify subpopulations of perivascular progenitor cells. Results showed that PDGFRα-expressing cells are present in four histologic niches within inguinal fat, including two perivascular locations. PDGFRα+ cells are most frequent within the tunica adventitia of arteries and veins, where PDGFRα+ cells populate the inner aspects of the adventitial layer. Although both PDGFRα+ and PDGFRα− fractions are multipotent progenitor cells, adipose tissue-derived PDGFRα+ stromal cells proliferate faster and mineralize to a greater degree than their PDGFRα− counterparts. Likewise, PDGFRα+ ectopic implants reconstitute the perivascular niche and ossify to a greater degree than PDGFRα− cell fractions. Adventicytes can be further grouped into three distinct groups based on expression of PDGFRα and/or CD34. When further partitioned, adventicytes co-expressing PDGFRα and CD34 represented a cell fraction with the highest mineralization potential. Long-term tracing studies showed that PDGFRα-expressing adventicytes give rise to adipocytes, but not to other cells within the vessel wall under homeostatic conditions. However, upon bone morphogenetic protein 2 (BMP2)-induced ossicle formation, descendants of PDGFRα+ cells gave rise to osteoblasts, adipocytes, and “pericyte-like” cells within the ossicle. In sum, PDGFRα marks distinct perivascular osteoprogenitor cell subpopulations within adipose tissue. The identification of perivascular osteoprogenitors may contribute to our improved understanding of pathologic mineralization/ossification.
- Published
- 2019
20. Comparison of Human Tissue Microarray to Human Pericyte Transcriptome Yields Novel Perivascular Cell Markers
- Author
-
Sarah Miller, Aaron W. James, Winters R. Hardy, Catherine Ding, Ching Yun Hsu, Bruno Péault, Carolyn A. Meyers, and Mario Gomez Salazar
- Subjects
0301 basic medicine ,Proteome ,Biology ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Original Research Reports ,Antigens, CD ,medicine ,Humans ,Perivascular Cell ,Progenitor cell ,Cells, Cultured ,Tissue microarray ,Gene Expression Profiling ,Mesenchymal stem cell ,Cell Biology ,Hematology ,Flow Cytometry ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Adipose Tissue ,Tissue Array Analysis ,Pericyte ,Pericytes ,Software ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Human perivascular progenitor cells, including pericytes, are well-described multipotent mesenchymal cells giving rise to mesenchymal stem cells in culture. Despite the unique location of pericytes, specific antigens to distinguish human pericytes from other cell types are few. Here, we employed a human tissue microarray (Human Protein Atlas) to identify proteins that are strongly and specifically expressed in a pericytic location within human adipose tissue. Next, these results were cross-referenced with RNA sequencing data from human adipose tissue pericytes, as defined as a fluorescence activated cell sorting (FACS) purified CD146(+)CD34(−)CD31(−)CD45(−) cell population. Results showed that from 105,532 core biopsies of soft tissue, 229 proteins showed strong and specific perivascular immunoreactivity, the majority of which (155) were present in the tunica intima. Next, cross-referencing with the transcriptome of FACS-derived CD146(+) pericytes yielded 25 consistently expressed genes/proteins, including 18 novel antigens. A majority of these transcripts showed maintained expression after culture propagation (56% of genes). Interestingly, many novel antigens within pericytes are regulators of osteogenic differentiation. In sum, our study demonstrates the existence of novel pericyte markers, some of which are conserved in culture that may be useful for future efforts to typify, isolate, and characterize human pericytes.
- Published
- 2019
21. Transplantation of human endometrial perivascular cells with elevated CYR61 expression induces angiogenesis and promotes repair of a full-thickness uterine injury in rat
- Author
-
Hairong Li, Xin’an Li, Zhongxun Li, Yong Liu, Qiang Diao, Haixiang Sun, Yimin Dai, Lijun Ding, Fei Yu, Bruno Péault, Yali Hu, Huaijun Zhou, Guijun Yan, Xiaoqiang Sheng, and Xin Zhen
- Subjects
Male ,0301 basic medicine ,Angiogenesis ,Uterus ,Fluorescent Antibody Technique ,Medicine (miscellaneous) ,Scars ,Endometrium ,Neovascularization ,0302 clinical medicine ,Pregnancy ,Tandem Mass Spectrometry ,lcsh:QD415-436 ,Cells, Cultured ,Endometrial perivascular cells ,lcsh:R5-920 ,Chemistry ,CYR61 ,Cell Differentiation ,Uterine horns ,Flow Cytometry ,Immunohistochemistry ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Myometrium ,Molecular Medicine ,Female ,Collagen ,medicine.symptom ,lcsh:Medicine (General) ,Neovascularization, Physiologic ,Enzyme-Linked Immunosorbent Assay ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Andrology ,lcsh:Biochemistry ,03 medical and health sciences ,medicine ,Animals ,Humans ,Regeneration ,Research ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Cell Biology ,Rats ,Transplantation ,030104 developmental biology ,Fertility ,Uterine injury ,Chromatography, Liquid ,Cysteine-Rich Protein 61 - Abstract
Background Disruptions of angiogenesis can have a significant effect on the healing of uterine scars. Human endometrial perivascular cells (CD146+PDGFRβ+) function as stem cells in the endometrium. Cysteine-rich angiogenic inducer 61 (CYR61) plays an important role in vascular development. The purpose of this study was to observe the effects of the transplantation of human endometrial perivascular cells (En-PSCs) overexpressing CYR61 on structural and functional regeneration in rat models of partial full-thickness uterine excision. Methods We first sorted human En-PSCs from endometrial single-cell suspensions by flow cytometry. Human En-PSCs expressing low or high levels of CYR61 were then generated via transfection with a CYR61-specific small interfering ribonucleic acid (si-CYR61) construct or overexpression plasmid. To establish a rat model of uterine injury, a subset of uterine wall was then resected from each uterine horn in experimental animals. Female rats were randomly assigned to five groups, including a sham-operated group and four repair groups that received either PBS loaded on a collagen scaffold (collagen/PBS), En-PSCs loaded on a collagen scaffold (collagen/En-PSCs), En-PSCs with low CYR61 expression loaded on a collagen scaffold (collagen/si-CYR61 En-PSCs), and En-PSCs overexpressing CYR61 loaded on a collagen scaffold (collagen/ov-CYR61 En-PSCs). These indicated constructs were sutured in the injured uterine area to replace the excised segment. On days 30 and 90 after transplantation, a subset of rats in each group was sacrificed, and uterine tissue was recovered and serially sectioned. Hematoxylin and eosin staining and immunohistochemical staining were then performed. Finally, the remaining rats of each group were mated with fertile male rats on day 90 for a 2-week period. Results Sorted En-PSCs expressed all recognized markers of mesenchymal stem cells (MSCs), including CD10, CD13, CD44, CD73, CD90, and CD105, and exhibited differentiation potential toward adipocytes, osteoblasts, and neuron-like cells. Compared with En-PSCs and En-PSCs with low CYR61 expression, En-PSCs with elevated CYR61 expression enhanced angiogenesis by in vitro co-culture assays. At day 90 after transplantation, blood vessel density in the collagen/ov-CYR61 En-PSCs group (11.667 ± 1.287) was greater than that in the collagen/En-PSCs group (7.167 ± 0.672) (P
- Published
- 2019
22. Stem Cells and Endometrial Regeneration: From Basic Research to Clinical Trial
- Author
-
Lijun Ding, Yali Hu, Guijun Yan, Bruno Péault, Xinxin Zhu, and Haixiang Sun
- Subjects
0301 basic medicine ,Biomedical Research ,Endometrial Cycle ,Medicine (miscellaneous) ,Asherman's syndrome ,Gynatresia ,Endometrium ,Bone marrow mesenchymal stem cells ,03 medical and health sciences ,0302 clinical medicine ,Basic research ,Animals ,Humans ,Regeneration ,Medicine ,Clinical Trials as Topic ,030219 obstetrics & reproductive medicine ,business.industry ,Regeneration (biology) ,Cell Differentiation ,Mesenchymal Stem Cells ,General Medicine ,medicine.disease ,Menstruation ,Clinical trial ,030104 developmental biology ,medicine.anatomical_structure ,Cancer research ,Female ,Stem cell ,business - Abstract
Monthly changes in the endometrial cycle indicate the presence of endometrial stem cells. In recent years, various stem cells that exist in the endometrium have been identified and characterized. Additionally, many studies have shown that Bone Marrow Mesenchymal Stem Cells (BM-MSCs) provide an alternative source for regenerating the endometrium and repairing endometrial injury. This review discusses the origin of endometrial stem cells, the characteristics and main biomarkers among five types of putative endometrial stem cells, applications of endometrium-derived stem cells and menstrual blood-derived stem cells, the association between BM-MSCs and endometrial stem cells, and progress in repairing endometrial injury.
- Published
- 2019
23. Ask the experts: current obstacles and future developments in cell culture
- Author
-
Bruno Péault, Daniel J. Kota, Jorge S. Burns, Justin A. Colacino, and Oren Ben-Yosef
- Subjects
Embryology ,Ask the Experts ,Ask price ,Stem Cells ,Political science ,Cell Culture Techniques ,Cell- and Tissue-Based Therapy ,Biomedical Engineering ,MEDLINE ,Humans ,Engineering ethics ,Current (fluid) ,Expert Testimony - Published
- 2019
24. Aging modulates the effects of ischemic injury upon mesenchymal cells within the renal interstitium and microvasculature
- Author
-
Eoin O'Sullivan, Kevin M Gallagher, David A. Ferenbach, Isaac Shaw, Gary Borthwick, Bruno Péault, Angela Oliveira Pisco, and Jeremy Hughes
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Medicine (General) ,kidney ,Aging ,Mesenchyme ,mesenchyme ,ischemia ,Interstitial cell ,03 medical and health sciences ,Mice ,0302 clinical medicine ,R5-920 ,pericyte ,Medicine ,Animals ,Humans ,Myofibroblasts ,Aged ,Kidney ,QH573-671 ,business.industry ,Mesenchymal stem cell ,fibrosis ,Cell Biology ,General Medicine ,030104 developmental biology ,medicine.anatomical_structure ,Tissue‐specific Progenitor and Stem cells ,Reperfusion Injury ,Microvessels ,CD146 ,Pericyte ,Wound healing ,business ,Cytology ,Pericytes ,Myofibroblast ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
The renal mesenchyme contains heterogeneous cells, including interstitial fibroblasts and pericytes, with key roles in wound healing. Although healing is impaired in aged kidneys, the effect of age and injury on the mesenchyme remains poorly understood. We characterized renal mesenchymal cell heterogeneity in young vs old animals and after ischemia‐reperfusion‐injury (IRI) using multiplex immunolabeling and single cell transcriptomics. Expression patterns of perivascular cell markers (α‐SMA, CD146, NG2, PDGFR‐α, and PDGFR‐β) correlated with their interstitial location. PDGFR‐α and PDGFR‐β co‐expression labeled renal myofibroblasts more efficiently than the current standard marker α‐SMA, and CD146 was a superior murine renal pericyte marker. Three renal mesenchymal subtypes; pericytes, fibroblasts, and myofibroblasts, were recapitulated with data from two independently performed single cell transcriptomic analyzes of murine kidneys, the first dataset an aging cohort and the second dataset injured kidneys following IRI. Mesenchymal cells segregated into subtypes with distinct patterns of expression with aging and following injury. Baseline uninjured old kidneys resembled post‐ischemic young kidneys, with this phenotype further exaggerated following IRI. These studies demonstrate that age modulates renal perivascular/interstitial cell marker expression and transcriptome at baseline and in response to injury and provide tools for the histological and transcriptomic analysis of renal mesenchymal cells, paving the way for more accurate classification of renal mesenchymal cell heterogeneity and identification of age‐specific pathways and targets., Kidney mesenchyme contains progenitor cells important in disease aetiology yet is poorly characterized with age and lacks research tools. Healthy and ischemia reperfusion injured (IRI) kidneys from old and young mice, analyzed using immunohistochemistry (IHC) and single cell RNA sequencing (scRNAseq), uncovered differences in aged injury response and provided more detailed characterization of kidney mesenchyme. BV = blood vessel.
- Published
- 2021
25. Microvascular maturation by mesenchymal stem cells in vitro improves blood perfusion in implanted tissue constructs
- Author
-
Rümeyza Bascetin, Anne Poliard, Clément Binet-Moussy, Catherine Chaussain, Philippe Girard, Laurent Muller, Stéphane Germain, Yoann Atlas, Caroline Gorin, Marion Marchand, Anita Novais, Bruno Péault, Sibylle Opsal-Vital, Catherine Monnot, Eirini Chatzopoulou, Julie Lesieur, Jérémy Sadoine, Matthieu Lesage, Centre interdisciplinaire de recherche en biologie (CIRB), Labex MemoLife, École normale supérieure - Paris (ENS Paris), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Ecole Superieure de Physique et de Chimie Industrielles de la Ville de Paris (ESPCI Paris), Université Paris sciences et lettres (PSL)-Collège de France (CdF (institution))-École normale supérieure - Paris (ENS Paris), Université Paris sciences et lettres (PSL)-Collège de France (CdF (institution))-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Sorbonne Université (SU), Laboratoires Pathologies, Imagerie et Biothérapies orofaciales, Partenaires INRAE, Hôpital Henri Mondor, University of California [Los Angeles] (UCLA), University of California, University of Edinburgh, Institut Jacques Monod (IJM (UMR_7592)), Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP), CCSD, Accord Elsevier, École normale supérieure - Paris (ENS-PSL), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Collège de France (CdF (institution))-Ecole Superieure de Physique et de Chimie Industrielles de la Ville de Paris (ESPCI Paris), Université Paris sciences et lettres (PSL)-École normale supérieure - Paris (ENS-PSL), Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Pathologies, Imagerie et Biothérapies oro-faciales (URP 2496), Université Paris Cité (UPCité), University of California (UC), Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité), Université Paris sciences et lettres (PSL)-Collège de France (CdF (institution))-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM), and Université de Paris (UP)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Basement membrane ,Angiogenesis ,[SDV]Life Sciences [q-bio] ,Cell ,Biophysics ,Dental pulp stem cells ,Mice, Nude ,Neovascularization, Physiologic ,Bioengineering ,02 engineering and technology ,Biology ,Vascular niche ,Perivascular recruitment ,Biomaterials ,Mice ,03 medical and health sciences ,medicine ,Animals ,Inosculation ,030304 developmental biology ,0303 health sciences ,Tissue Engineering ,Mesenchymal stem cell ,Endothelial Cells ,Mesenchymal Stem Cells ,021001 nanoscience & nanotechnology ,Cell biology ,In vitro maturation ,Perfusion ,[SDV] Life Sciences [q-bio] ,medicine.anatomical_structure ,Mechanics of Materials ,Ceramics and Composites ,Stem cell ,0210 nano-technology - Abstract
International audience; Blood perfusion of grafted tissue constructs is a hindrance to the success of stem cell-based therapies by limiting cell survival and tissue regeneration. Implantation of a pre-vascularized network engineered in vitro has thus emerged as a promising strategy for promoting blood supply deep into the construct, relying on inosculation with the host vasculature. We aimed to fabricate in vitro tissue constructs with mature microvascular networks, displaying perivascular recruitment and basement membrane, taking advantage of the angiogenic properties of dental pulp stem cells and self-assembly of endothelial cells into capillaries. Using digital scanned light-sheet microscopy, we characterized the generation of dense microvascular networks in collagen hydrogels and established parameters for quantification of perivascular recruitment. We also performed original time-lapse analysis of stem cell recruitment. These experiments demonstrated that perivascular recruitment of dental pulp stem cells is driven by PDGF-BB. Recruited stem cells participated in deposition of vascular basement membrane and vessel maturation. Mature microvascular networks thus generated were then compared to those lacking perivascular coverage generated using stem cell conditioned medium. Implantation in athymic nude mice demonstrated that in vitro maturation of microvascular networks improved blood perfusion and cell survival within the construct. Taken together, these data demonstrate the strong potential of in vitro production of mature microvasculature for improving cell-based therapies.
- Published
- 2021
26. Bone-forming perivascular cells: Cellular heterogeneity and use for tissue repair
- Author
-
Jiajia Xu, Stefano Negri, Yiyun Wang, Winters R. Hardy, Ginny Ching Yun Hsu, Lijun Ding, Aaron W. James, Bruno Péault, Mario Gomez-Salazar, and Zhao Li
- Subjects
Gene isoform ,ALDH ,Biology ,LAMP1 ,Article ,adipogenesis ,osteogenesis ,CD107a ,Antigen ,In vivo ,perivascular stem cell ,Animals ,Progenitor cell ,mesenchymal stem cell ,Mammals ,Wound Healing ,Tissue Engineering ,tunica adventitia ,Tunica Adventitia ,Mesenchymal stem cell ,adipose stem cell ,Cell Differentiation ,Mesenchymal Stem Cells ,Cell Biology ,Cell biology ,Adipogenesis ,mesenchymal stromal cell ,Molecular Medicine ,CD10 ,Pericytes ,exocytosis ,Developmental Biology ,CD140a - Abstract
Mesenchymal progenitor cells are broadly distributed across perivascular niches—an observation conserved between species. One common histologic zone with a high frequency of mesenchymal progenitor cells within mammalian tissues is the tunica adventitia, the outer layer of blood vessel walls populated by cells with a fibroblastic morphology. The diversity and functions of (re)generative cells present in this outermost perivascular niche are under intense investigation; we have reviewed herein our current knowledge of adventitial cell potential with a somewhat narrow focus on bone formation. Antigens of interest to functionally segregate adventicytes are discussed, including CD10, CD107a, aldehyde dehydrogenase isoforms, and CD140a, among others. Purified adventicytes (such as CD10+, CD107alow, and CD140a+ cells) have stronger osteogenic potential and promote bone formation in vivo. Recent bone tissue engineering applications of adventitial cells are also presented. A better understanding of perivascular progenitor cell subsets may represent a beneficial advance for future efforts in tissue repair and bioengineering.
- Published
- 2021
27. Role of Pericytes in the Development of the Renin/Angiotensin System: Induction of Functional Renin in Cultures of Pericytes
- Author
-
Bruno Péault, Nusrat Khan, Ania Stefanska, John J. Mullins, Christopher J. Kenyon, and Angela Briski
- Subjects
0301 basic medicine ,Kidney ,Primary culture ,Angiogenesis ,030204 cardiovascular system & hematology ,Biology ,Cell recruitment ,medicine.disease ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Fibrosis ,Renin–angiotensin system ,medicine ,Secretion - Abstract
Renal pericytes have a critical importance for angiogenesis and vascular remodeling, medullary blood flow regulation, and development of fibrosis. An emerging role for kidney pericytes is their ability to induce renin expression and synthesis. Here, we present methods for purification of human renal pericytes, their primary culture, and differentiation into renin-producing cells. Possible applications of these protocols include investigations into (1) renin cell recruitment mechanisms, (2) modulation of renin expression/secretion by small molecules, and (3) renin expression/secretion in nonrenal pericytes. A potential therapeutic application of this work is the identification of new players regulating the renin-angiotensin system.
- Published
- 2021
28. Assessing the Bone-Forming Potential of Pericytes
- Author
-
Hsin Chuan Pan, Jiajia Xu, Kang Ting, Carolyn A. Meyers, Jia Shen, Bruno Péault, Aaron W. James, Chenchao Wang, and Chia Soo
- Subjects
0301 basic medicine ,education.field_of_study ,Cell type ,Mesenchymal stem cell ,Population ,Biology ,Cell sorting ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Tissue engineering ,medicine ,Pericyte ,Stem cell ,Bone regeneration ,education ,030217 neurology & neurosurgery - Abstract
Human pericytes are a perivascular cell population with mesenchymal stem cell properties, present in all vascularized tissues. Human pericytes have a distinct immunoprofile, which may be leveraged for purposes of cell purification. Adipose tissue is the most commonly used cell source for human pericyte derivation. Pericytes can be isolated by FACS (fluorescence-activated cell sorting), most commonly procured from liposuction aspirates. Pericytes have clonal multilineage differentiation potential, and their potential utility for bone regeneration has been described across multiple animal models. The following review will discuss in vivo methods for assessing the bone-forming potential of purified pericytes. Potential models include (1) mouse intramuscular implantation, (2) mouse calvarial defect implantation, and (3) rat spinal fusion models. In addition, the presented surgical protocols may be used for the in vivo analysis of other osteoprogenitor cell types.
- Published
- 2021
29. Divergent effects of distinct perivascular cell subsets for intra-articular cell therapy in posttraumatic osteoarthritis
- Author
-
Stefano Negri, Yiyun Wang, Jiajia Xu, Bruno Péault, Masnsen Cherief, Aaron W. James, Ginny Ching Yun Hsu, and Takashi Sono
- Subjects
Cartilage, Articular ,Stromal cell ,Receptor, Platelet-Derived Growth Factor alpha ,Intra-Articular ,0206 medical engineering ,Cell ,Cell- and Tissue-Based Therapy ,Chondrocyte hypertrophy ,synovium ,Mice, Transgenic ,02 engineering and technology ,White adipose tissue ,Osteoarthritis ,Inbred C57BL ,Article ,Transgenic ,Injections, Intra-Articular ,Injections ,Cell therapy ,03 medical and health sciences ,Mice ,0302 clinical medicine ,adventitial cell ,medicine ,Animals ,Orthopedics and Sports Medicine ,Perivascular space ,DMM ,030203 arthritis & rheumatology ,Sclerosis ,Chemistry ,Animal ,Mesenchymal stem cell ,Platelet-Derived Growth Factor alpha ,medicine.disease ,020601 biomedical engineering ,Pdgfrβ ,Mice, Inbred C57BL ,Disease Models, Animal ,medicine.anatomical_structure ,Cartilage ,Disease Models ,Cancer research ,Pdgfrα ,cell therapy ,Receptor ,Articular - Abstract
Intra-articular injection of mesenchymal stem cells has shown benefit for the treatment of osteoarthritis (OA). However, mesenchymal stem/stromal cells at the origin of these clinical results are heterogenous cell populations with limited cellular characterization. Here, two transgenic reporter mice were used to examine the differential effects of two precisely defined perivascular cell populations (Pdgfrα+ and Pdgfrβ+ cells) from white adipose tissue for alleviation of OA. Perivascular mesenchymal cells were isolated from transgenic Pdgfrα-and Pdgfrβ-CreERT2 reporter animals and delivered as a one-time intra-articular dose to C57BL/6J mice after destabilization of the medial meniscus (DMM). Both Pdgfrα+ and Pdgfrβ+ cell preparations improved metrics of cartilage degradation and reduced markers of chondrocyte hypertrophy. While some similarities in cell distribution were identified within the synovial and perivascular spaces, injected Pdgfrα+ cells remained in the superficial layers of articular cartilage, while Pdgfrβ+ cells were more widely dispersed. Pdgfrβ+ cell therapy prevented subchondral sclerosis induced by DMM, while Pdgfrα+ cell therapy had no effect. In summary, while both cell therapies showed beneficial effects in the DMM model, important differences in cell incorporation, persistence, and subchondral sclerosis were identified.
- Published
- 2021
30. Author response: Lysosomal protein surface expression discriminates fat- from bone-forming human mesenchymal precursor cells
- Author
-
Stefano Negri, Ching Yun Hsu, Yongxing Gao, Kristen P. Broderick, Leslie Chang, Yiyun Wang, Ye Tian, Nusrat Kahn, Winters R. Hardy, Carolyn A. Meyers, Robert J. Tower, Bruno Péault, Shuaishuai Hu, Aaron W. James, Takashi Sono, and Jiajia Xu
- Subjects
Chemistry ,Precursor cell ,Mesenchymal stem cell ,Bone forming ,Surface protein ,Cell biology - Published
- 2020
31. Comparison of skeletal and soft tissue pericytes identifies CXCR4+ bone forming mural cells in human tissues
- Author
-
Jody E. Hooper, Leititia Zhang, Yongxing Gao, Leslie Chang, Sridhar Nimmagadda, Yiyun Wang, Aaron W. James, Jiajia Xu, Carol D. Morris, Dongqing Li, Ching Yun Hsu, Kristen P. Broderick, Carolyn A. Meyers, Ye Tian, Bruno Péault, and Robert J. Tower
- Subjects
0301 basic medicine ,Histology ,Stromal cell ,Physiology ,Endocrinology, Diabetes and Metabolism ,Adipose tissue ,Diseases ,lcsh:Physiology ,Mural cell ,Article ,03 medical and health sciences ,0302 clinical medicine ,Precursor cell ,medicine ,Progenitor cell ,Bone ,lcsh:QH301-705.5 ,lcsh:QP1-981 ,Chemistry ,Mesenchymal stem cell ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Biology (General) ,CD146 ,Pericyte ,030217 neurology & neurosurgery - Abstract
Human osteogenic progenitors are not precisely defined, being primarily studied as heterogeneous multipotent cell populations and termed mesenchymal stem cells (MSCs). Notably, select human pericytes can develop into bone-forming osteoblasts. Here, we sought to define the differentiation potential of CD146+ human pericytes from skeletal and soft tissue sources, with the underlying goal of defining cell surface markers that typify an osteoblastogenic pericyte. CD146+CD31−CD45− pericytes were derived by fluorescence-activated cell sorting from human periosteum, adipose, or dermal tissue. Periosteal CD146+CD31−CD45− cells retained canonical features of pericytes/MSC. Periosteal pericytes demonstrated a striking tendency to undergo osteoblastogenesis in vitro and skeletogenesis in vivo, while soft tissue pericytes did not readily. Transcriptome analysis revealed higher CXCR4 signaling among periosteal pericytes in comparison to their soft tissue counterparts, and CXCR4 chemical inhibition abrogated ectopic ossification by periosteal pericytes. Conversely, enrichment of CXCR4+ pericytes or stromal cells identified an osteoblastic/non-adipocytic precursor cell. In sum, human skeletal and soft tissue pericytes differ in their basal abilities to form bone. Diversity exists in soft tissue pericytes, however, and CXCR4+ pericytes represent an osteoblastogenic, non-adipocytic cell precursor. Indeed, enrichment for CXCR4-expressing stromal cells is a potential new tactic for skeletal tissue engineering.
- Published
- 2020
32. Platelet Derived Growth Factor Receptor-β (PDGFRβ) lineage tracing highlights perivascular cell to myofibroblast transdifferentiation during post-traumatic osteoarthritis
- Author
-
Stefano Negri, Ching Yun Hsu, Jiajia Xu, Bruno Péault, Carolyn A. Meyers, Takashi Sono, Sarah Miller, Yiyun Wang, and Aaron W. James
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Angiogenesis ,PDGFRβ ,0206 medical engineering ,Mice, Transgenic ,02 engineering and technology ,Transgenic ,Article ,Receptor, Platelet-Derived Growth Factor beta ,Experimental ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Vasculogenesis ,Growth factor receptor ,Genes, Reporter ,pericyte ,Osteoarthritis ,perivascular stem cell ,medicine ,Animals ,Orthopedics and Sports Medicine ,Cell Lineage ,Myofibroblasts ,Reporter ,DMM ,030203 arthritis & rheumatology ,biology ,Arthritis ,Transdifferentiation ,020601 biomedical engineering ,Arthritis, Experimental ,Fibrosis ,myofibroblast ,Platelet-Derived Growth Factor beta ,medicine.anatomical_structure ,Genes ,Cell Transdifferentiation ,biology.protein ,Female ,Joints ,Pericyte ,Pericytes ,Myofibroblast ,Platelet-derived growth factor receptor ,Receptor - Abstract
Pericytes ubiquitously surround capillaries and microvessels within vascularized tissues and have diverse functions after tissue injury. In addition to regulation of angiogenesis and tissue regeneration after injury, pericytes also contribute to organ fibrosis. Destabilization of the medial meniscus (DMM) phenocopies post-traumatic osteoarthritis, yet little is known regarding the impact of DMM surgery on knee joint-associated pericytes and their cellular descendants. Here, inducible platelet-derived growth factor receptor-β (PDGFRβ)-CreERT2 reporter mice were subjected to DMM surgery, and lineage tracing studies performed over an 8-week period. Results showed that at baseline PDGFRβ reporter activity highlights abluminal perivascular cells within synovial and infrapatellar fat pad (IFP) tissues. DMM induces a temporospatially patterned increase in vascular density within synovial and subsynovial tissues. Marked vasculogenesis within IFP was accompanied by expansion of PDGFRβ reporter+ perivascular cell numbers, detachment of mGFP+ descendants from vessel walls, and aberrant adoption of myofibroblastic markers among mGFP+ cells including α-SMA, ED-A, and TGF-β1. At later timepoints, fibrotic changes and vascular maturation occurred within subsynovial tissues, with the redistribution of PDGFRβ+ cellular descendants back to their perivascular niche. In sum, PDGFRβ lineage tracing allows for tracing of perivascular cell fate within the diarthrodial joint. Further, destabilization of the joint induces vascular and fibrogenic changes of the IFP accompanied by perivascular to myofibroblast transdifferentiation.
- Published
- 2020
33. Perivascular Fibro-Adipogenic Progenitor Tracing during Post-Traumatic Osteoarthritis
- Author
-
Takashi Sono, Jiajia Xu, Amanda K. Huber, Benjamin Levi, Simone Marini, Masnsen Cherief, Sarah Miller, Ching Yun Hsu, Bruno Péault, Aaron W. James, and Yiyun Wang
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Knee Joint ,Mice, Transgenic ,PDGFRA ,Fat pad ,Mural cell ,Article ,Pathology and Forensic Medicine ,Green fluorescent protein ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Fibrosis ,Gene expression ,Osteoarthritis ,medicine ,Adipocytes ,Animals ,Cell Lineage ,Progenitor cell ,Infrapatellar fat pad ,business.industry ,Stem Cells ,Fibroblasts ,medicine.disease ,030104 developmental biology ,Adipose Tissue ,business ,030217 neurology & neurosurgery - Abstract
Perivascular mural cells surround capillaries and microvessels and have diverse regenerative or fibrotic functions after tissue injury. Subsynovial fibrosis is a well-known pathologic feature of osteoarthritis, yet transgenic animals for use in visualizing perivascular cell contribution to fibrosis during arthritic changes have not been developed. Here, inducible Pdgfra-CreER(T2) reporter mice were subjected to joint-destabilization surgery to induce arthritic changes, and cell lineage was traced over an 8-week period with a focus on the joint-associated fat pad. Results showed that, at baseline, inducible Pdgfra reporter activity highlighted adventitial and, to a lesser extent, pericytic cells within the infrapatellar fat pad. Joint-destabilization surgery was associated with marked fibrosis of the infrapatellar fat pad, accompanied by an expansion of perivascular Pdgfra-expressing cellular descendants, many of which adopted α-smooth muscle actin expression. Gene expression analysis of microdissected infrapatellar fat pad confirmed enrichment in membrane-bound green fluorescent protein/Pdgfra-expressing cells, along with a gene signature that corresponded with injury-associated fibro-adipogenic progenitors. Our results highlight dynamic changes in joint-associated perivascular fibro-adipogenic progenitors during osteoarthritis.
- Published
- 2020
34. WISP-1 drives bone formation at the expense of fat formation in human perivascular stem cells
- Author
-
Bruno Péault, Kang Ting, Greg Asatrian, Jia Shen, Chia Soo, Aaron W. James, Catherine Ding, Jiajia Xu, Kristen P. Broderick, and Carolyn A. Meyers
- Subjects
0301 basic medicine ,Cell type ,Stromal cell ,Science ,Population ,Cell Separation ,Biology ,Bone and Bones ,Article ,Adipogenic Differentiation Conditions ,CCN Intercellular Signaling Proteins ,Fats ,03 medical and health sciences ,Osteogenesis ,Proto-Oncogene Proteins ,Humans ,Perivascular Niche ,Progenitor cell ,education ,Cells, Cultured ,Osteogenic Differentiation ,education.field_of_study ,Multidisciplinary ,Tissue Engineering ,Stem Cells ,Matricellular protein ,Mesenchymal stem cell ,Cell Differentiation ,Flow Cytometry ,FACS Purification ,Cell biology ,Up-Regulation ,Transplantation ,Segmental Spinal Fusion ,030104 developmental biology ,Adipose Tissue ,Cellular Microenvironment ,Medicine ,Stem cell ,Pericytes - Abstract
The vascular wall within adipose tissue is a source of mesenchymal progenitors, referred to as perivascular stem/stromal cells (PSC). PSC are isolated via fluorescence activated cell sorting (FACS), and defined as a bipartite population of pericytes and adventitial progenitor cells (APCs). Those factors that promote the differentiation of PSC into bone or fat cell types are not well understood. Here, we observed high expression of WISP-1 among human PSC in vivo, after purification, and upon transplantation in a bone defect. Next, modulation of WISP-1 expression was performed, using WISP-1 overexpression, WISP-1 protein, or WISP-1 siRNA. Results demonstrated that WISP-1 is expressed in the perivascular niche, and high expression is maintained after purification of PSC, and upon transplantation in a bone microenvironment. In vitro studies demonstrate that WISP-1 has pro-osteogenic/anti-adipocytic effects in human PSC, and that regulation of BMP signaling activity may underlie these effects. In summary, our results demonstrate the importance of the matricellular protein WISP-1 in regulation of the differentiation of human stem cell types within the perivascular niche. WISP-1 signaling upregulation may be of future benefit in cell therapy mediated bone tissue engineering, for the healing of bone defects or other orthopaedic applications.
- Published
- 2018
35. Neer Award 2018: Platelet-derived growth factor receptor α co-expression typifies a subset of platelet-derived growth factor receptor β–positive progenitor cells that contribute to fatty degeneration and fibrosis of the murine rotator cuff
- Author
-
Sai K. Devana, Allison Ariniello, Brandon K. Vu, Bruno Péault, Ayelet Dar, Frank A. Petrigliano, Gina M Mosich, Andrew R. Jensen, Paras Shah, Claire D. Eliasberg, Benjamin V Kelley, and Iain R. Murray
- Subjects
0301 basic medicine ,Receptor, Platelet-Derived Growth Factor alpha ,Muscle Fibers, Skeletal ,Cell ,Awards and Prizes ,Mice, Transgenic ,Rotator Cuff Injuries ,Green fluorescent protein ,Receptor, Platelet-Derived Growth Factor beta ,Mice ,Rotator Cuff ,03 medical and health sciences ,Growth factor receptor ,Fibrosis ,medicine ,Animals ,Orthopedics and Sports Medicine ,Progenitor cell ,Cells, Cultured ,Adipogenesis ,biology ,business.industry ,Stem Cells ,General Medicine ,medicine.disease ,In vitro ,Tissue Degeneration ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Adipose Tissue ,Cancer research ,biology.protein ,Surgery ,Atrophy ,business ,Platelet-derived growth factor receptor - Abstract
Background and hypothesis After massive tears, rotator cuff muscle often undergoes atrophy, fibrosis, and fatty degeneration. These changes can lead to high surgical failure rates and poor patient outcomes. The identity of the progenitor cells involved in these processes has not been fully elucidated. Platelet-derived growth factor receptor β (PDGFRβ) and platelet-derived growth factor receptor α (PDGFRα) have previously been recognized as markers of cells involved in muscle fibroadipogenesis. We hypothesized that PDGFRα expression identifies a fibroadipogenic subset of PDGFRβ+ progenitor cells that contribute to fibroadipogenesis of the rotator cuff. Methods We created massive rotator cuff tears in a transgenic strain of mice that allows PDGFRβ+ cells to be tracked via green fluorescent protein (GFP) fluorescence. We then harvested rotator cuff muscle tissues at multiple time points postoperatively and analyzed them for the presence and localization of GFP+ PDGFRβ+ PDGFRα+ cells. We cultured, induced, and treated these cells with the molecular inhibitor CWHM-12 to assess fibrosis inhibition. Results GFP+ PDGFRβ+ PDGFRα+ cells were present in rotator cuff muscle tissue and, after massive tears, localized to fibrotic and adipogenic tissues. The frequency of PDGFRβ+ PDGFRα+ cells increased at 5 days after massive cuff tears and decreased to basal levels within 2 weeks. PDGFRβ+ PDGFRα+ cells were highly adipogenic and significantly more fibrogenic than PDGFRβ+ PDGFRα– cells in vitro and localized to adipogenic and fibrotic tissues in vivo. Treatment with CWHM-12 significantly decreased fibrogenesis from PDGFRβ+ PDGFRα+ cells. Conclusion PDGFRβ+ PDGFRα+ cells directly contribute to fibrosis and fatty degeneration after massive rotator cuff tears in the mouse model. In addition, CWHM-12 treatment inhibits fibrogenesis from PDGFRβ+ PDGFRα+ cells in vitro. Clinically, perioperative PDGFRβ+ PDGFRα+ cell inhibition may limit rotator cuff tissue degeneration and, ultimately, improve surgical outcomes for massive rotator cuff tears.
- Published
- 2018
36. Pericytes in the renal vasculature: roles in health and disease
- Author
-
Jeremy Hughes, Isaac Shaw, Bruno Péault, John J. Mullins, and Sebastien A. Rider
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Stromal cell ,Angiogenesis ,Neovascularization, Physiologic ,Blood Pressure ,Kidney ,urologic and male genital diseases ,03 medical and health sciences ,0302 clinical medicine ,Renin–angiotensin system ,medicine ,Renal fibrosis ,Animals ,Humans ,Cell Lineage ,Zebrafish ,business.industry ,Mesenchymal stem cell ,Glomerulosclerosis ,Mesenchymal Stem Cells ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Nephrology ,Blood Vessels ,Kidney Diseases ,Pericytes ,business ,030217 neurology & neurosurgery ,Homeostasis - Abstract
In the dense circulatory system of the kidney, as in all vascularized tissues, pericytes enwrap capillaries and microvessels to regulate angiogenesis, stabilize microvascular networks and control blood flow by vasoconstriction. Specialized renal pericytes known as mesangial cells provide physical support to glomerular capillaries, whereas a subset of juxtaglomerular arteriolar pericytes control the local blood pressure in the glomerulus via contraction and influence systemic blood pressure by secreting renin. Similar to pericytes from many other organs, cultured human renal pericytes give rise to mesenchymal stem/stromal cells, suggesting a role of perivascular cells in renal homeostasis and regeneration. On the other hand, pericytes directly contribute to renal fibrosis, and mesangial cells may have an essential role in the development of glomerulosclerosis and other nephropathies. From their early emergence in the renal embryonic rudiment to their distribution in diverse perivascular niches in the adult organ, we review the anatomy and function of pericytes in the healthy and diseased kidney. Many aspects of the ontogeny, specification and functional specialization of renal pericytes remain elusive. The development of powerful models in the easily accessible and genetically tractable zebrafish will help to uncover the multiple facets of these cells.
- Published
- 2018
37. Early Immunomodulatory Effects of Implanted Human Perivascular Stromal Cells During Bone Formation
- Author
-
Jiajia Xu, Bruno Péault, Justin M. Sacks, Chia Soo, Xinli Zhang, Aaron W. James, Kristen P. Broderick, Kang Ting, Noah Yan, Greg Asatrian, Lei Zhang, Catherine Ding, Raghav Goyal, and Carolyn A. Meyers
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Stromal cell ,endocrine system diseases ,Population ,Biomedical Engineering ,Bone Matrix ,Adipose tissue ,Bioengineering ,Mice, SCID ,Biology ,Mesenchymal Stem Cell Transplantation ,digestive system ,Biochemistry ,Immunomodulation ,Biomaterials ,Mice ,03 medical and health sciences ,Osteogenesis ,medicine ,Animals ,Humans ,Bone formation ,Progenitor cell ,education ,education.field_of_study ,digestive, oral, and skin physiology ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Original Articles ,Cells, Immobilized ,digestive system diseases ,030104 developmental biology ,Cytokines ,Heterografts - Abstract
Human perivascular stem/stromal cells (PSC) are a multipotent mesodermal progenitor cell population defined by their perivascular residence. PSC are most commonly derived from subcutaneous adipose tissue, and recent studies have demonstrated the high potential for clinical translation of this fluorescence-activated cell sorting-derived cell population for bone tissue engineering. Specifically, purified PSC induce greater bone formation than unpurified stroma taken from the same patient sample. In this study, we examined the differences in early innate immune response to human PSC or unpurified stroma (stromal vascular fraction [SVF]) during the in vivo process of bone formation. Briefly, SVF or PSC from the same patient sample were implanted intramuscularly in the hindlimb of severe combined immunodeficient (SCID) mice using an osteoinductive demineralized bone matrix carrier. Histological examination of early inflammatory infiltrates was examined by hematoxylin and eosin and immunohistochemical staining (Ly-6G, F4/80). Results showed significantly greater neutrophilic and macrophage infiltrates within and around SVF in comparison to PSC-laden implants. Differences in early postoperative inflammation among SVF-laden implants were associated with reduced osteogenic differentiation and bone formation. Similar findings were recapitulated with PSC implantation in immunocompetent mice. Exaggerated postoperative inflammation was associated with increased IL-1α, IL-1β, IFN-γ, and TNF-α gene expression among SVF samples, and conversely increased IL-6 and IL-10 expression among PSC samples. These data document a robust immunomodulatory effect of implanted PSC, and an inverse correlation between host inflammatory cell infiltration and stromal progenitor cell-mediated ossification.
- Published
- 2018
38. Transcriptional Networks in Single Perivascular Cells Sorted from Human Adipose Tissue Reveal a Hierarchy of Mesenchymal Stem Cells
- Author
-
W. Reef Hardy, Chirayu P. Goswami, Leni Moldovan, Krishnalekha Datta, Bruno Péault, Mirko Corselli, Iain R. Murray, Keith L. March, Kenneth J. Livak, Dmitry O. Traktuev, and Nicanor I. Moldovan
- Subjects
0301 basic medicine ,Stromal cell ,CD34 ,Biology ,03 medical and health sciences ,Humans ,Cell Lineage ,Gene Regulatory Networks ,Progenitor cell ,Stem cell transplantation for articular cartilage repair ,Mesenchymal stem cell ,Cell Differentiation ,Mesenchymal Stem Cells ,Cell Biology ,Aldehyde Dehydrogenase ,Middle Aged ,Stromal vascular fraction ,Flow Cytometry ,Cell biology ,030104 developmental biology ,Adipose Tissue ,Gene Expression Regulation ,Immunology ,Molecular Medicine ,CD146 ,Female ,Single-Cell Analysis ,Stem cell ,Pericytes ,Developmental Biology - Abstract
Adipose tissue is a rich source of multipotent mesenchymal stem-like cells, located in the perivascular niche. Based on their surface markers, these have been assigned to two main categories: CD31−/CD45−/CD34+/CD146− cells (adventitial stromal/stem cells [ASCs]) and CD31−/CD45−/CD34−/CD146+ cells (pericytes [PCs]). These populations display heterogeneity of unknown significance. We hypothesized that aldehyde dehydrogenase (ALDH) activity, a functional marker of primitivity, could help to better define ASC and PC subclasses. To this end, the stromal vascular fraction from a human lipoaspirate was simultaneously stained with fluorescent antibodies to CD31, CD45, CD34, and CD146 antigens and the ALDH substrate Aldefluor, then sorted by fluorescence-activated cell sorting. Individual ASCs (n = 67) and PCs (n = 73) selected from the extremities of the ALDH-staining spectrum were transcriptionally profiled by Fluidigm single-cell quantitative polymerase chain reaction for a predefined set (n = 429) of marker genes. To these single-cell data, we applied differential expression and principal component and clustering analysis, as well as an original gene coexpression network reconstruction algorithm. Despite the stochasticity at the single-cell level, covariation of gene expression analysis yielded multiple network connectivity parameters suggesting that these perivascular progenitor cell subclasses possess the following order of maturity: (a) ALDHbrASC (most primitive); (b) ALDHdimASC; (c) ALDHbrPC; (d) ALDHdimPC (least primitive). This order was independently supported by specific combinations of class-specific expressed genes and further confirmed by the analysis of associated signaling pathways. In conclusion, single-cell transcriptional analysis of four populations isolated from fat by surface markers and enzyme activity suggests a developmental hierarchy among perivascular mesenchymal stem cells supported by markers and coexpression networks.
- Published
- 2017
39. Pericytes for the treatment of orthopedic conditions
- Author
-
Iain R. Murray, Bruno Péault, Tulyapruek Tawonsawatruk, Greg Asatrian, Hindle P, Christopher C. West, Vi Nguyen, Jia Shen, Aaron W. James, Xinli Zhang, Kang Ting, A Hamish R W Simpson, and Chia Soo
- Subjects
0301 basic medicine ,Stromal cell ,Angiogenesis ,Population ,Rodentia ,Biology ,Bone and Bones ,Article ,03 medical and health sciences ,medicine ,Animals ,Humans ,Pharmacology (medical) ,Musculoskeletal Diseases ,education ,Pharmacology ,education.field_of_study ,Tissue Engineering ,Stem Cells ,Mesenchymal stem cell ,Cell Differentiation ,Mesenchymal Stem Cells ,Cell biology ,Transplantation ,Cartilage ,030104 developmental biology ,medicine.anatomical_structure ,Immunology ,CD146 ,Pericyte ,Stem cell ,Pericytes - Abstract
Pericytes and other perivascular stem cells are of growing interest in orthopedics and tissue engineering. Long regarded as simple regulators of angiogenesis and blood pressure, pericytes are now recognized to have MSC (mesenchymal stem cell) characteristics, including multipotentiality, self-renewal, immunoregulatory functions, and diverse roles in tissue repair. Pericytes are typified by characteristic cell surface marker expression (including αSMA, CD146, PDGFRβ, NG2, RGS5, among others). Although alone no marker is absolutely specific for pericytes, collectively these markers appear to selectively identify an MSC-like pericyte. The purification of pericytes is most well described as a CD146(+)CD34(−)CD45(−) cell population. Pericytes and other perivascular stem cell populations have been applied in diverse orthopedic applications, including both ectopic and orthotopic models. Application of purified cells has sped calvarial repair, induced spine fusion, and prevented fibrous non-union in rodent models. Pericytes induce these effects via both direct and indirect mechanisms. In terms of their paracrine effects, pericytes are known to produce and secrete high levels of a number of growth and differentiation factors both in vitro and after transplantation. The following review will cover existing studies to date regarding pericyte application for bone and cartilage engineering. In addition, further questions in the field will be pondered, including the phenotypic and functional overlap between pericytes and culture-derived MSC, and the concept of pericytes as efficient producers of differentiation factors to speed tissue repair.
- Published
- 2017
40. Human Adipose Tissue Micro-fragmentation for Cell Phenotyping and Secretome Characterization
- Author
-
Bruno Péault, Carlo Tremolada, Mario Gomez-Salazar, Bianca Vezzani, and Joan Casamitjana
- Subjects
0301 basic medicine ,Stromal vascular fraction ,Cells ,General Chemical Engineering ,Adipose tissue ,Biology ,Mesenchymal Stem Cell Transplantation ,General Biochemistry, Genetics and Molecular Biology ,NO ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Adipocyte ,medicine ,Autologous transplantation ,Humans ,Cells, Cultured ,Mesenchymal stem cell ,Pericyte ,Cultured ,General Immunology and Microbiology ,Regeneration (biology) ,General Neuroscience ,Cell biology ,Transplantation ,030104 developmental biology ,medicine.anatomical_structure ,Phenotype ,chemistry ,Adipose Tissue ,Micro-fragmentation ,Adventitial cell ,Cytokines ,Intercellular Signaling Peptides and Proteins ,Pericytes ,030217 neurology & neurosurgery - Abstract
In the past decade, adipose tissue transplants have been widely used in plastic surgery and orthopaedics to enhance tissue repletion and/or regeneration. Accordingly, techniques for harvesting and processing human adipose tissue have evolved in order to quickly and efficiently obtain large amounts of tissue. Among these, the closed system technology represents an innovative and easy-to-use system to harvest, process, and re-inject refined fat tissue in a short time and in the same intervention (intra-operatively). Adipose tissue is collected by liposuction, washed, emulsified, rinsed and minced mechanically into cell clusters of 0.3 to 0.8 mm. Autologous transplantation of mechanically fragmented adipose tissue has shown remarkable efficacy in different therapeutic indications such as aesthetic medicine and surgery, orthopedic and general surgery. Characterization of micro-fragmented adipose tissue revealed the presence of intact small vessels within the adipocyte clusters; hence, the perivascular niche is left unperturbed. These clusters are enriched in perivascular cells (i.e., mesenchymal stem cell (MSC) ancestors) and in vitro analysis showed an increased release of growth factors and cytokines involved in tissue repair and regeneration, compared to enzymatically derived MSCs. This suggests that the superior therapeutic potential of microfragmented adipose tissue is explained by a higher frequency of presumptive MSCs and enhanced secretory activity. Whether these added pericytes directly contribute to higher growth factor and cytokine production is not known. This clinically approved procedure allows the transplantation of presumptive MSCs without the need for expansion and/or enzymatic treatment, thus bypassing the requirements of GMP guidelines, and reducing the costs for cell-based therapies.
- Published
- 2019
41. Human Adipose-derived Pericytes Display Steroidogenic Lineage Potential in Vitro and Influence Leydig Cell Regeneration in Vivo in Rats
- Author
-
Laura Milne, Ian Handel, Lee B. Smith, Michael Curley, Z. Gonzalez, Patrick W. F. Hadoke, and Bruno Péault
- Subjects
Male ,medicine.medical_specialty ,medicine.drug_class ,Adipose tissue ,lcsh:Medicine ,Reproductive biology ,Cell Count ,Biology ,Rats, Inbred WKY ,Article ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Internal medicine ,Testis ,medicine ,Regeneration ,Animals ,Humans ,Cell Lineage ,RNA, Messenger ,lcsh:Science ,Testosterone ,Cells, Cultured ,030304 developmental biology ,0303 health sciences ,030219 obstetrics & reproductive medicine ,Multidisciplinary ,Leydig cell ,Regeneration (biology) ,lcsh:R ,Leydig Cells ,Organ Size ,Androgen ,Hormones ,Transplantation ,medicine.anatomical_structure ,Endocrinology ,Adipose Tissue ,Gene Expression Regulation ,lcsh:Q ,Steroids ,Stem cell ,Pericytes - Abstract
Exogenous androgen replacement is used to treat symptoms associated with low testosterone in males. However, adverse cardiovascular risk and negative fertility impacts impel development of alternative approaches to restore/maintain Leydig cell (LC) androgen production. Stem Leydig cell (SLC) transplantation shows promise in this regard however, practicality of SLC isolation/transplantation impede clinical translation. Multipotent human adipose-derived perivascular stem cells (hAd-PSCs) represent an attractive extragonadal stem cell source for regenerative therapies in the testis but their therapeutic potential in this context is unexplored. We asked whether hAd-PSCs could be converted into Leydig-like cells and determined their capacity to promote regeneration in LC-ablated rat testes. Exposure of hAd-PSCs to differentiation-inducing factors in vitro upregulated steroidogenic genes but did not fully induce LC differentiation. In vivo, no difference in LC-regeneration was noted between Sham and hAd-PSC-transplanted rats. Interestingly, Cyp17a1 expression increased in hAd-PSC-transplanted testes compared to intact vehicle controls and the luteinising hormone/testosterone ratio returned to Vehicle control levels which was not the case in EDS + Sham animals. Notably, hAd-PSCs were undetectable one-month after transplantation suggesting this effect is likely mediated via paracrine mechanisms during the initial stages of regeneration; either directly by interacting with regenerating LCs, or through indirect interactions with trophic macrophages.
- Published
- 2019
42. Aged Mice Demonstrate Greater Muscle Degeneration of Chronically Injured Rotator Cuff
- Author
-
Bruno Péault, Jonathan D Gatto, Allison Ariniello, Paras Shah, Frank A. Petrigliano, Gina M Mosich, Abhinav K Sharma, David R. McAllister, Vivian J Hu, Regina Husman, Ayelet Dar, Andrew R. Jensen, Brandon Levian, and Daniel J McClintick
- Subjects
Cell type ,Pathology ,medicine.medical_specialty ,Aging ,Adipose tissue ,Rotator Cuff Injuries ,Fibrosis ,medicine ,Animals ,Humans ,Orthopedics and Sports Medicine ,Rotator cuff ,Progenitor cell ,Adiposity ,Aged ,business.industry ,Age Factors ,Skeletal muscle ,Middle Aged ,medicine.disease ,Muscle atrophy ,Mice, Inbred C57BL ,Muscular Atrophy ,medicine.anatomical_structure ,Tears ,medicine.symptom ,business - Abstract
Massive tears of the rotator cuff (RC) are often associated with progressive and irreversible muscle degeneration due to fibrosis, fatty infiltration, and muscle atrophy. RC tears are common in individuals older than 60 years and the repair of these tears is amongst the most prevalent of orthopedic procedures. However, most current models of this injury are established in young animals, which may not accurately recapitulate the clinical condition. In this study, we used a murine model of massive RC tears to evaluate age-related muscle degeneration following chronic injury. The expression of the fibro-adipogenic genes encoding collagen type III and leptin was higher in aged RC compared with matched injured young tissue at 2 weeks post-injury, and development of fibrosis was accelerated in aged mice within 5 days post-injury. Furthermore, the synthesis of collagens type I and III and fat tissue accumulation were significantly higher in injured RCs of aged mice. Similar frequency of fibro-adipogenic PDGFRβ+ PDGFRα+ progenitor cells was measured in non-injured RC of aged and young mice, but PDGFRβ+ PDGFRα+ cells contributed to significantly larger fibrotic lesions in aged RCs within 2 weeks post-injury, implying a more robust fibrotic environment in the aged injured muscle. Altogether, these findings demonstrate age-dependent differences in RC response to chronic injury with a more profound fibro-adipogenic change in aged muscles. Clinically, cell therapies for muscular pathologies should not only consider the cell type being transplanted but also the recipient milieu into which these cells are seeded. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:320-328, 2020.
- Published
- 2019
43. Human perivascular stem cell-derived extracellular vesicles mediate bone repair
- Author
-
Bruno Péault, Carolyn A. Meyers, Kenneth W. Witwer, Yiyun Wang, Kristen P. Broderick, Ching Yun Hsu, Yongxing Gao, Aaron W. James, Jiajia Xu, Leslie Chang, Catherine Ding, and Leititia Zhang
- Subjects
0301 basic medicine ,Mouse ,QH301-705.5 ,Science ,Exosome ,Osteocytes ,General Biochemistry, Genetics and Molecular Biology ,osteogenesis ,03 medical and health sciences ,Paracrine signalling ,Extracellular Vesicles ,0302 clinical medicine ,pericyte ,medicine ,Humans ,exosome ,Progenitor cell ,Biology (General) ,bone tissue engineering ,Cells, Cultured ,mesenchymal stem cell ,General Immunology and Microbiology ,Chemistry ,General Neuroscience ,Regeneration (biology) ,Stem Cells ,Mesenchymal stem cell ,General Medicine ,Cell Biology ,bone repair ,Stem Cells and Regenerative Medicine ,Coculture Techniques ,Trypsinization ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Blood Vessels ,Medicine ,Pericyte ,Stem cell ,030217 neurology & neurosurgery ,Research Article ,Human - Abstract
The vascular wall is a source of progenitor cells that are able to induce skeletal repair, primarily by paracrine mechanisms. Here, the paracrine role of extracellular vesicles (EVs) in bone healing was investigated. First, purified human perivascular stem cells (PSCs) were observed to induce mitogenic, pro-migratory, and pro-osteogenic effects on osteoprogenitor cells while in non-contact co-culture via elaboration of EVs. PSC-derived EVs shared mitogenic, pro-migratory, and pro-osteogenic properties of their parent cell. PSC-EV effects were dependent on surface-associated tetraspanins, as demonstrated by EV trypsinization, or neutralizing antibodies for CD9 or CD81. Moreover, shRNA knockdown in recipient cells demonstrated requirement for the CD9/CD81 binding partners IGSF8 and PTGFRN for EV bioactivity. Finally, PSC-EVs stimulated bone repair, and did so via stimulation of skeletal cell proliferation, migration, and osteodifferentiation. In sum, PSC-EVs mediate the same tissue repair effects of perivascular stem cells, and represent an ‘off-the-shelf’ alternative for bone tissue regeneration., eLife digest Throughout our lives, our bodies need to heal after injury. Blood vessels are found throughout the body’s tissues and are a source of cells that guide the process of repair. These cells, called perivascular stem cells (PSCs), are a type of stem cell found in the lining of blood vessels. Stem cells are cells that can become one of several different types of mature cells, depending on what the body needs. Extracellular vesicles are bundles of chemical signals that cells send into their external environment. Just like an address or a tag on a parcel, specific molecules mark the exterior surface of these bundles to deliver the message to the right recipient. Stem cells often use extracellular vesicles to communicate with surrounding cells. One role of PSCs is repairing damage to bones. Unusually, they do not turn into new bone cells and so do not directly contribute to the re-growing tissue. Instead, PSCs act indirectly, by stimulating the cells around them. How PSCs send these ‘repair instructions’ has, however, remained unclear. Xu et al. wanted to determine if PSCs used extracellular vesicles to direct bone repair, and if so, what ‘tags’ needed to be on the vesicles and on the receiving cells for this to happen. Experiments using PSCs and immature bone cells grown in the laboratory allowed the PSCs’ effect on bone cells to be simulated in a Petri dish. The two types of cells were grown on either side of a barrier, which separated them physically but allowed chemical signals through. In response to the PSCs, the immature bone cells multiplied, started to move (which is something they need to do to heal damaged tissue), and began to resemble mature bone cells. Analysis of the signals released by the PSCs revealed that these were indeed extracellular vesicles, and that they were tagged by specific proteins called tetraspanins. Genetic manipulation of the immature bone cells later showed that these cells needed specific ‘receiver’ molecules to respond to the PSCs. Adding only extracellular vesicles to the bone cells, without any PSCs, confirmed that it was indeed the vesicles that triggered the healing response. Finally, giving the vesicles to mice with bone damage helped them to heal faster than untreated animals. These results have uncovered a key mechanism by which stem cells control the repair of bone tissue. This could one day lead to better treatments for patients recovering from fractures or needing bone surgery.
- Published
- 2019
44. Author response: Human perivascular stem cell-derived extracellular vesicles mediate bone repair
- Author
-
Leslie Chang, Yiyun Wang, Carolyn A. Meyers, Kenneth W. Witwer, Aaron W. James, Jiajia Xu, Yongxing Gao, Kristen P. Broderick, Leititia Zhang, Bruno Péault, Ching Yun Hsu, and Catherine Ding
- Subjects
Chemistry ,Bone healing ,Stem cell ,Extracellular vesicles ,Cell biology - Published
- 2019
45. International Expert Consensus on a Cell Therapy Communication Tool: DOSES
- Author
-
Aaron J. Krych, Rodrigo Mardones, Jason L. Dragoo, Bert R. Mandelbaum, Christian Lattermann, Norimasa Nakamura, Lars Engebretsen, Henning Madry, Arnold I. Caplan, Daniel B.F. Saris, Frank A. Petrigliano, Johnny Huard, Andrew G. Geeslin, Robert F. LaPrade, Denis Evseenko, Jorge Chahla, Elizaveta Kon, A Hamish R W Simpson, Scott A. Rodeo, Alan Getgood, Marc R. Safran, Nicola Maffulli, James H.-C. Wang, Mark A. Birch, Chris Hyunchul Jo, Constance R. Chu, L. C. Biant, Allan B. Dietz, Matthew J. Dalby, Bruno Péault, Farshid Guilak, Brian J. Cole, Anthony P. Hollander, and Iain R. Murray
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Scientific Articles ,Consensus ,Standardization ,Delphi Technique ,Arthroplasty, Replacement, Hip ,Delphi method ,MEDLINE ,Cell- and Tissue-Based Therapy ,Prosthesis Design ,03 medical and health sciences ,0302 clinical medicine ,Basic research ,Terminology as Topic ,Medicine and Health Sciences ,Medicine ,Humans ,Orthopedics and Sports Medicine ,Medical physics ,030222 orthopedics ,business.industry ,Communication ,Expert consensus ,General Medicine ,Reference Standards ,Transparency (behavior) ,030104 developmental biology ,Surgery ,business - Abstract
© 2019 BY THE JOURNAL OF BONE AND JOINT SURGERY, INCORPORATED. Background:The lack of a standardized system for describing cell therapies acts as a barrier to advancement in clinical and basic research and practice. The aim of this study was to establish an international expert consensus on strategies to improve standardization and transparency when describing cell therapies. The secondary aim was to develop a consensus among experts on the contents of a standardized tool for describing cell therapies.Methods:The need for expert consensus on strategies to improve cell therapy communication was confirmed at the American Academy of Orthopaedic Surgeons/National Institutes of Health Optimizing Clinical Use of Biologics Symposium in 2018. A working group of 6 experts convened an international consensus process involving clinicians and basic scientists using validated Delphi methodology. This iterative process was used to define statements on communication of cell therapies and develop a standardized tool for describing cell therapies.Results:Thirty-four experts completed 3 rounds survey with use of the Delphi process. After 3 rounds, 27 statements relating to existing nomenclature, solutions to improve communication, ideal characteristics of a framework, mandatory elements of a new framework, and future work to facilitate application reached consensus with >80% agreement and
- Published
- 2019
46. CD10 expression identifies a subset of human perivascular progenitor cells with high proliferation and calcification potentials
- Author
-
Lu Xu, Bruno Péault, Zhongzhou Yang, Yong Liu, Lijun Ding, Yali Hu, W. Reef Hardy, Nusrat Khan, Anushri Gaur, Jing Su, Bianca Vezzani, Guijun Yan, Haixiang Sun, Zhenyu Diao, Aaron W. James, and Ruotian Li
- Subjects
0301 basic medicine ,Adult ,GLI1 ,Cell ,Cell fate determination ,SHH ,NO ,osteogenesis ,adventitia ,03 medical and health sciences ,0302 clinical medicine ,Calcification, Physiologic ,immune system diseases ,hemic and lymphatic diseases ,Adventitia ,medicine ,Humans ,Progenitor cell ,neoplasms ,mesenchymal stem cell ,Aged ,Cell Proliferation ,Aged, 80 and over ,biology ,Stem Cells ,Mesenchymal stem cell ,Cell Biology ,Cell cycle ,Middle Aged ,medicine.disease ,adventitia, CD10, GLI1, osteogenesis, mesenchymal stem cell, SHH, vascular calcification ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,vascular calcification ,biology.protein ,CD10 ,Molecular Medicine ,Neprilysin ,030217 neurology & neurosurgery ,Developmental Biology ,Calcification - Abstract
The tunica adventitia ensheathes arteries and veins and contains presumptive mesenchymal stem cells (MSCs) involved in vascular remodeling. We show here that a subset of human adventitial cells express the CD10/CALLA cell surface metalloprotease. Both CD10+ and CD10− adventitial cells displayed phenotypic features of MSCs when expanded in culture. However, CD10+ adventitial cells exhibited higher proliferation, clonogenic and osteogenic potentials in comparison to their CD10− counterparts. CD10+ adventitial cells increased expression of the cell cycle protein CCND2 via ERK1/2 signaling and osteoblastogenic gene expression via NF-κB signaling. CD10 expression was upregulated in adventitial cells through sonic hedgehog-mediated GLI1 signaling. These results suggest that CD10, which marks rapidly dividing cells in other normal and malignant cell lineages, plays a role in perivascular MSC function and cell fate specification. These findings also point to a role for CD10+ perivascular cells in vascular remodeling and calcification. Significance statement Perivascular adventitial cells include multipotent progenitor cells giving rise in culture to mesenchymal stem/progenitor cells. The present data show that a subset of human adventitial cells natively express the CD10 surface marker, regulated by sonic hedgehog/GLI1 signaling. Purified CD10+ adventitial cells exhibit high proliferative, clonogenic and osteogenic potentials, suggesting a role in pathologic vascular remodeling.
- Published
- 2019
47. Mesenchymal stem cell perspective: cell biology to clinical progress
- Author
-
Dennis E. Discher, Mark F. Pittenger, Donald G. Phinney, Arnold I. Caplan, Joshua M. Hare, and Bruno Péault
- Subjects
0301 basic medicine ,Stromal cell ,Cell ,Population ,Biomedical Engineering ,lcsh:Medicine ,Medicine (miscellaneous) ,Review Article ,Biology ,Cell therapy ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Progenitor cell ,education ,education.field_of_study ,lcsh:R ,Mesenchymal stem cell ,Cell Biology ,Human cell ,Stem-cell research ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Mesenchymal stem cells ,Expansive ,Neuroscience ,Developmental Biology - Abstract
The terms MSC and MSCs have become the preferred acronym to describe a cell and a cell population of multipotential stem/progenitor cells commonly referred to as mesenchymal stem cells, multipotential stromal cells, mesenchymal stromal cells, and mesenchymal progenitor cells. The MSCs can differentiate to important lineages under defined conditions in vitro and in limited situations after implantation in vivo. MSCs were isolated and described about 30 years ago and now there are over 55,000 publications on MSCs readily available. Here, we have focused on human MSCs whenever possible. The MSCs have broad anti-inflammatory and immune-modulatory properties. At present, these provide the greatest focus of human MSCs in clinical testing; however, the properties of cultured MSCs in vitro suggest they can have broader applications. The medical utility of MSCs continues to be investigated in over 950 clinical trials. There has been much progress in understanding MSCs over the years, and there is a strong foundation for future scientific research and clinical applications, but also some important questions remain to be answered. Developing further methods to understand and unlock MSC potential through intracellular and intercellular signaling, biomedical engineering, delivery methods and patient selection should all provide substantial advancements in the coming years and greater clinical opportunities. The expansive and growing field of MSC research is teaching us basic human cell biology as well as how to use this type of cell for cellular therapy in a variety of clinical settings, and while much promise is evident, careful new work is still needed.
- Published
- 2019
48. Relative contributions of adipose-resident CD146 pericytes and CD34 adventitial progenitor cells in bone tissue engineering
- Author
-
Carolyn A. Meyers, Yiyun Wang, Aaron W. James, Lei Zhang, Jiajia Xu, Kristen P. Broderick, Leslie Chang, Min Lee, and Bruno Péault
- Subjects
0301 basic medicine ,Cell type ,Stromal cell ,Angiogenesis ,Chemistry ,Mesenchymal stem cell ,lcsh:R ,Biomedical Engineering ,CD34 ,Medicine (miscellaneous) ,Connective tissue ,lcsh:Medicine ,Cell Biology ,Article ,3. Good health ,Cell biology ,Cell therapy ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,medicine ,Progenitor cell ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Pericytes and other perivascular stem/stromal cells are of growing interest in the field of tissue engineering. A portion of perivascular cells are well recognized to have MSC (mesenchymal stem cell) characteristics, including multipotentiality, self-renewal, immunoregulatory functions, and diverse roles in tissue repair. Here, we investigate the differential but overlapping roles of two perivascular cell subsets in paracrine induction of bone repair. CD146+CD34−CD31−CD45−pericytes and CD34+CD146−CD31−CD45−adventitial cells were derived from human adipose tissue and applied alone or in combination to calvarial bone defects in mice. In vitro, osteogenic differentiation and tubulogenesis assays were performed using either fluorescence activated cell sorting-derived CD146+ pericytes or CD34+ adventitial cells. Results showed that CD146+ pericytes induced increased cord formation in vitro and angiogenesis in vivo in comparison with patient-matched CD34+ adventitial cells. In contrast, CD34+ adventitial cells demonstrated heightened paracrine-induced osteogenesis in vitro. When applied in a critical-size calvarial defect model in NOD/SCID mice, the combination treatment of CD146+ pericytes with CD34+ adventitial cells led to greater re-ossification than either cell type alone. In summary, adipose-derived CD146+ pericytes and CD34+ adventitial cells display functionally distinct yet overlapping and complementary roles in bone defect repair. Consequently, CD146+ pericytes and CD34+ adventitial cells may demonstrate synergistic bone healing when applied as a combination cellular therapy., Bone repair: synergistic healing from two types of fat cells Different kinds of cells found surrounding blood vessels in fat play a complementary and synergistic role in bone healing. Aaron James from Johns Hopkins University in Baltimore, MD, USA, and colleagues derived two subsets of cells from human fat tissue: contractile cells known as pericytes that wrap around cellular lining of capillaries and tiny veins; and connective tissue cells known as adventitial cells that surrounds larger vessels. Under isolated culture conditions, pericytes stimulated the development of primitive blood vessels, whereas adventitial cells promoted early bone formation. The researchers applied the cells to the sites of bone defects in mice and saw that combined treatment with both pericytes and adventitial cells led to greater bone repair than treatment with either cell type alone.
- Published
- 2019
49. Perivascular Progenitor Cells for Bone Regeneration
- Author
-
Winters R. Hardy, Noah Yan, Kang Ting, Aaron W. James, Hindle P, Bruno Péault, Chia Soo, Jia Jia Xu, Carolyn A. Meyers, Greg Asatrian, and Kristen P. Broderick
- Subjects
education.field_of_study ,Pathology ,medicine.medical_specialty ,Stromal cell ,Regeneration (biology) ,Mesenchymal stem cell ,Population ,Biology ,medicine.anatomical_structure ,medicine ,Pericyte ,Progenitor cell ,Bone regeneration ,education ,Multipotentiality - Abstract
Perivascular progenitor cells are of growing interest in the field of bone tissue engineering. Perivascular progenitor cells have mesenchymal stem/stromal cell (MSC) characteristics, including multipotentiality, self-renewal, immunomodulatory functions, and diverse roles in tissue repair. From human tissue, the purification of perivascular progenitor cells is most common from subcutaneous white adipose tissue, although all vascularized organs studied to date have a perivascular progenitor cell population. Microvascular pericytes are commonly isolated as a CD146+CD34−CD31−CD45− cell population, while adventitial progenitor cells are more commonly identified as a CD146−CD34+CD31−CD45− population. Perivascular progenitor cells have been applied in diverse orthopedic conditions, including both ectopic and orthotopic models of bone formation/regeneration. This review covers studies to date in bone tissue engineering as well as several emerging areas of study, including the concept of regional specification within the perivascular niche.
- Published
- 2019
50. FOXP1 circular RNA sustains mesenchymal stem cell identity via microRNA inhibition
- Author
-
Mario Barilani, Lorenza Lazzari, Hamish Simpson, Riccardo L. Rossi, G. Buono, Giovanna Cantarella, Francesco Rusconi, Enrico Ragni, Murtadhah M K Jalal, Bruno Péault, and Alessandro Cherubini
- Subjects
Pluripotent Stem Cells ,Cytoplasm ,Cellular differentiation ,Biology ,Immunophenotyping ,Mesoderm ,03 medical and health sciences ,0302 clinical medicine ,Circular RNA ,microRNA ,RNA and RNA-protein complexes ,Genetics ,Humans ,Gene Silencing ,RNA, Small Interfering ,Induced pluripotent stem cell ,Cell Proliferation ,Oligonucleotide Array Sequence Analysis ,030304 developmental biology ,Cell Nucleus ,0303 health sciences ,Sequence Analysis, RNA ,Gene Expression Profiling ,Stem Cells ,Mesenchymal stem cell ,Wnt signaling pathway ,Cell Differentiation ,Forkhead Transcription Factors ,Mesenchymal Stem Cells ,RNA, Circular ,Fibroblasts ,Cell biology ,ErbB Receptors ,Repressor Proteins ,Wnt Proteins ,MicroRNAs ,HEK293 Cells ,030220 oncology & carcinogenesis ,Exoribonucleases ,RNA ,Stem cell ,Reprogramming - Abstract
Stem cell identity and plasticity are controlled by master regulatory genes and complex circuits also involving non-coding RNAs. Circular RNAs (circRNAs) are a class of RNAs generated from protein-coding genes by backsplicing, resulting in stable RNA structures devoid of free 5’ and 3’ ends. Little is known of the mechanisms of action of circRNAs, let alone in stem cell biology. In this study, for the first time, we determined that a circRNA controls mesenchymal stem cell (MSC) identity and differentiation. High-throughput MSC expression profiling from different tissues revealed a large number of expressed circRNAs. Among those, circFOXP1 was enriched in MSCs compared to differentiated mesodermal derivatives. Silencing of circFOXP1 dramatically impaired MSC differentiation in culture and in vivo. Furthermore, we demonstrated a direct interaction between circFOXP1 and miR-17–3p/miR-127–5p, which results in the modulation of non-canonical Wnt and EGFR pathways. Finally, we addressed the interplay between canonical and non-canonical Wnt pathways. Reprogramming to pluripotency of MSCs reduced circFOXP1 and non-canonical Wnt, whereas canonical Wnt was boosted. The opposing effect was observed during generation of MSCs from human pluripotent stem cells. Our results provide unprecedented evidence for a regulatory role for circFOXP1 as a gatekeeper of pivotal stem cell molecular networks.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.