Editorial Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Stem Cells. May 26, 2024; 16(5): 479-485
Published online May 26, 2024. doi: 10.4252/wjsc.v16.i5.479
Deer antler stem cell niche: An interesting perspective
Claudia Cavallini, Carlo Ventura, National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems - Eldor Lab, Bologna 40128, Italy
Claudia Cavallini, Elena Olivi, Riccardo Tassinari, Chiara Zannini, Gregorio Ragazzini, Valentina Taglioli, Eldor Lab, Bologna 40128, Italy
Martina Marcuzzi, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna 40138, Italy
ORCID number: Claudia Cavallini (0000-0002-8079-9697); Elena Olivi (0000-0002-8999-1538); Riccardo Tassinari (0000-0002-9425-1841); Chiara Zannini (0000-0003-3925-4844); Gregorio Ragazzini (0000-0002-6876-5080); Martina Marcuzzi (0000-0002-8965-3168); Valentina Taglioli (0000-0002-0687-9576); Carlo Ventura (0000-0001-9333-0321).
Author contributions: Cavallini C, Olivi E, and Ventura C contributed to this work with literature review and analysis; Tassinari R, Zannini C, Ragazzini G, Marcuzzi M, and Taglioli V contributed to the discussion and design of the manuscript; and all the authors contributed to this paper with drafting, critical revision and editing, and approval of the final version.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Carlo Ventura, MD, PhD, Director, Full Professor, National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems - Eldor Lab, Via Corticella 183, Bologna 40128, Italy. carlo.ventura@unibo.it
Received: February 21, 2024
Revised: April 9, 2024
Accepted: April 25, 2024
Published online: May 26, 2024
Processing time: 92 Days and 21.1 Hours

Abstract

In recent years, there has been considerable exploration into methods aimed at enhancing the regenerative capacity of transplanted and/or tissue-resident cells. Biomaterials, in particular, have garnered significant interest for their potential to serve as natural scaffolds for cells. In this editorial, we provide commentary on the study by Wang et al, in a recently published issue of World J Stem Cells, which investigates the use of a decellularized xenogeneic extracellular matrix (ECM) derived from antler stem cells for repairing osteochondral defects in rat knee joints. Our focus lies specifically on the crucial role of biological scaffolds as a strategy for augmenting stem cell potential and regenerative capabilities, thanks to the establishment of a favorable microenvironment (niche). Stem cell differentiation heavily depends on exposure to intrinsic properties of the ECM, including its chemical and protein composition, as well as the mechanical forces it can generate. Collectively, these physicochemical cues contribute to a bio-instructive signaling environment that offers tissue-specific guidance for achieving effective repair and regeneration. The interest in mechanobiology, often conceptualized as a form of “structural memory”, is steadily gaining more validation and momentum, especially in light of findings such as these.

Key Words: Extracellular matrix, Antler stem cells, Stem cell niche, Regenerative medicine, Decellularized scaffolds, Cell memory

Core Tip: Recent research has focused on enhancing cell regenerative capacity through biomaterials, particularly natural scaffolds. A novel study published in World J Stem Cells investigates the possibility of using a decellularized xenogeneic extracellular matrix (ECM) from antler stem cells to repair osteochondral defects in rats. Our editorial emphasizes the vital role of biological scaffolds in boosting stem cell potential and regenerative abilities by creating a favorable microenvironment. Stem cell differentiation relies on the ECM properties, including its chemical composition and mechanical forces. This bio-instructive signaling environment offers tissue-specific guidance for effective repair and regeneration, aligning with the growing interest in mechanobiology.



INTRODUCTION

In recent years, new and intriguing strategies aimed at enhancing the regenerative capabilities of stem cells during transplantation have been emerging. Among these, of particular interest are biomaterials capable of providing natural scaffolds to the transplanted, as well as (tissue) resident cells. In this editorial, we comment on the article that appeared in a recent issue of World J Stem Cells titled “High quality repair of osteochondral defects in rats using the extracellular matrix of antler stem cells”[1], in relation to other recent papers on similar topics. Furthermore, we aim to draw inspiration from this interesting subject for broader reflections, regarding stem cell niches and morphogenesis.

Adult stem cells, of mesenchymal origin in particular, have proven to be a promising possibility in the field of regenerative medicine, given their ability to differentiate into different cell types, as well as their ability to secrete trophic factors capable of modulating the trophism of recipient tissues, additionally showcasing a relevant immunomodulatory potential[2]. Many clinical trials are currently underway worldwide, and at the time of writing there are about 1500 clinical trials underway (https://clinicaltrials.gov/).

Despite the great potential, the beneficial effect derived from stem cell transplantation, mesenchymal stem cells (MSCs) in particular, appears to be moderate. Indeed, unmodified MSCs have shown limited advantages in various preclinical and clinical investigations, primarily attributable to challenges related to differences between MSCs derived from variegated sources, variability in culturing protocols, and, after transplantation, different engraftment efficiencies, inconsistent cell homing as well as cell viability[3]. All these issues prompted extensive research efforts over the past few decades to enhance cell functionality and potency.

In the past years, there has been exploration into different approaches to enhance the regenerative capabilities of stem cells. Various strategies have been tried, including cytokines and growth factors, hypoxia, pharmacological drugs, biomaterials, various culture conditions, a variety of biologically active molecules, and many others[4]. In this context, the utilization of scaffolds is particularly intriguing. The topic is of particular interest, as evidenced by the results of the PubMed search for the words “stem cell AND extracellular matrix”, that leads to 18010 results. However, if we circumvent the search by entering the words “animal-derived AND extracellular matrix” the results go down to 52, and they drop even further if keywords are “antler stem cell AND extracellular matrix”, that lead to 8 results, showing that this is still an under-explored field. Due to the limited number of publications, it is not only easier to grasp the state of the art, but we also consider this a clear indication of the necessity to delve into these emerging research domains.

BIOLOGICAL SCAFFOLD: A STRATEGY TO IMPROVE STEM CELL POTENTIAL

Considering the underwhelming outcomes in stem cell differentiation, it is reasonable to surmise that something pertaining specifically to the differentiation process might have been overlooked. The interaction between stem cells and their surrounding microenvironment plays a fundamental role in numerous processes, including cell migration, proliferation, lineage specificity, and tissue morphogenesis, thanks to the establishment of a conducive niche for the optimal expression of stem cell capabilities[5,6].

In vivo, the fate determination of stem cells is intricately regulated by a complex array of signals orchestrated within the cellular microenvironment. Within the stem cell niche, cell-to-cell signaling interactions are regulated by different components: Soluble factors from surrounding cells and/or tissues, the extracellular matrix (ECM) or cell substrate, the biophysical milieu, etc[7]. The sophisticated control mechanisms evolved by organisms to manage cell populations, including stem cells, are still largely unknown. Despite current gaps of knowledge, the importance of these structures remains unquestionably crucial.

Scaffolds not only can serve as platforms for in vitro cell growth before transplantation, but even more fascinating may be their direct transplantation in vivo, where they can serve as support for endogenous stem cells, which would go on to repopulate this type of structures. Indeed, a supportive microenvironment might be essential for regulating stem cell function, thereby activating or enhancing intrinsic host repair mechanisms[8]. There are several methods to investigate the chemical composition of the matrix: Biochemical analysis, scanning electron microscopy, transmission electron microscopy, infrared spectroscopy, raman spectroscopy, mass spectrometry, etc. Conversely, it remains much more difficult to study the mechanical properties, as well as the spatial interaction of structure proteins with colonizing cells. Despite this, it is clear that the mechano-sensitive pathways translate physical cues into biochemical signals, directing the cell towards a particular lineage[9].

Numerous efforts have been undertaken to comprehensively identify and characterize the influence of specific environmental cues on stem cell behavior, in order to achieve the most optimized support for cell growth and differentiation. Additionally, various materials, including both synthetic and natural ones, have been developed and, in many instances, already subjected to testing[10]. The inherent physicochemical characteristics of the biomaterial play a crucial role in cell behavior. Relevant properties include matrix stiffness[11-13], matrix porosity[14-16], topography, such as roughness and patterns[17-19], viscoelasticity[20,21], hydrophobicity[22], and surface charge[23]. Biomaterials used as support for cell growth are of different types, whether natural or synthetic, manufactured with different kinds of materials, such as ceramics, wood, and plastic, used alone or combined with living cells and tissues, also derived from animals[24].

Natural scaffolds are typically obtained through decellularization procedures, which are performed to eliminate cells and their components, particularly DNA and RNA, from the ECM. This process yields a natural matrix with preserved mechanical integrity[25]. Decellularized ECM (dECM) has been shown to be a viable type of natural scaffold for tissue engineering, as the ECM plays a critical role in tissue development. Studies for the possible use of decellularized scaffolds have been conducted since the 1950s[26]. In particular, the advent of dECM scaffolds offers a promising avenue in regenerative medicine, emulating an optimal non-immune environment, featuring native three-dimensional structures and a diverse array of bioactive components[27]. Decellularized scaffolds can be classified in several ways, one of which involves categorizing them into scaffolds derived from tissues and organs via decellularization, and scaffolds derived from matrix deposition by cells[28]. dECM scaffolds have been studied and applied in regenerative medicine for the repair or replacement of various tissues, e.g., skin, bone, heart, nerves, liver, lung, and kidney[27].

Drawing upon studies focused on the heart, an organ of particular interest in regenerative medicine due to the profound health implications of heart disease and the limited regenerative potential of its constituent cells, numerous investigations have been conducted since the 1990s. As early as 1999, a study documented the transplantation of decellularized pig-derived valves into sheep, revealing promising in vivo recellularization alongside the absence of calcification, a prognostically negative factor[29]. In the years that ensued, a multitude of studies have been published on this topic, collectively showcasing its feasibility, albeit without widespread adoption in clinical practice. This is due to issues regarding in vitro decellularization methods, as well as in vivo or ex vivo recellularization strategies[30].

An example of an interesting biomaterial derived from animal source is mentioned in the article we are taking into account. A field of possible scaffold massive clinical application is orthopedics, where prosthetics is widely used. The utilization of scaffolds capable of replacing or restoring damaged tissues holds great significance. It is precisely within this domain that biomaterials of various origins are under thorough investigation[31].

TISSUE SPECIFICITY OF ECM

This short roundup shows how stem cell fate is shaped by numerous factors and tangled interactions, through orchestrated engagement with soluble factors, neighboring cells, and ECMs: A localized biochemical and mechanical environment is established, characterized by intricate and dynamic regulatory patterning that stem cells perceive[32]. It is now clear that the capacity of stem cells to initiate differentiation into mature tissue cells is dependent on exposure to intrinsic properties of the ECM, determined by the chemical and protein composition, but also by the nature of the mechanical forces that the matrix is capable of generating[9], influenced, at least partially, by the origin of ECM itself.

Compelling experimental evidence has shown that ECM produced, even in vitro, by cells of different origins has very different properties. For example, ECM produced by stromal cells derived from human bone marrow (BM) and human adipose tissue (AD), BM-ECM and AD-ECM respectively, shows better abilities to induce proliferation in stem cells of equal origin, that is BM cells for the former and adipose derived cells for the latter. Additionally, to indicate further origin-dependent specificity, BM- and AD-ECM were found to selectively guide human MSC differentiation towards either osteogenic or adipogenic lineages, respectively, indicating tissue-specific effects of the ECM[33]. Even more interesting, is that ECM influenced cell morphology regardless of the human MSC origin, further supporting the notion of tissue specificity in the observed effects.

It is thus evident how the structures that specific cell types form are provided with a kind of memory (retained by the cells themselves), which also influences the cells that will later colonize those structures. In the same way, decellularized organs were shown to be substrates capable of promoting differentiation in the cell types they naturally harbored. The bio-instructive signaling cues found in dECM-based grafts can offer tissue-specific guidance for directing cellular behavior and coordinating cellular chemotaxis, as demonstrated in various applications: Regeneration of skeletal muscle[34], liver[35], trachea[36], and many others. There are already commercially available decellularized tissue-based products approved for clinical use, and, for instance, among these, heart valves and xenogeneic grafts made from bovine carotid arteries are the most commonly used[37].

Scientific evidence thus seems to suggest that ECM derived from cells with specific characteristics, including cancer, exhibits distinct properties. Much literature has paid attention regarding changes in ECM during cancer progression, demonstrating that dysregulation of ECM composition, structure, stiffness, and abundance contribute to invasiveness[38]. More intriguingly, ECM derived from healthy cells has the potential to yield beneficial effects in diseased contexts. ECM derived from human MSCs did not promote the proliferation of the cancer cell line HeLa, MCF-7, and MDA-MB-231[33]. When breast tumor cell lines, exhibiting various levels of invasiveness (benign, non-invasive, and invasive), were cultured in their own ECM or in ECM deposited by the other cell lines, they showed distinct cellular responses, correlating with the malignancy of the cell sources utilized in the ECM preparation. Accordingly, ECM derived from normal mammary gland cells were found to inhibit breast cancer proliferation[39]. As well, dECM obtained from normal lung tissue prompted apoptosis in MCF-7 cells, also inhibiting epithelial-mesenchymal transition, a common feature of malignancy[40].

THE IMPORTANCE OF ORIGIN

With the aim of identifying and studying mechanisms capable of inducing rapid tissue regeneration, the peculiar nature of deer antlers is of particular interest, as addressed in the commented article. Deer antlers are organs distinguishing them from other mammals, showing an incredible speed of growth, where rapid cell proliferation is elegantly controlled without resulting into malignancy[41]. Because of this feature, antler stem cells (ASCs) can serve as a model for examining the proteins and pathways implicated in maintaining a stem cell niche, as well as their activation and differentiation during organogenesis.

Animal derivatives are largely used in traditional medicine all over the world. The World Health Organization estimates that up to 80% of the global population (numbering over six billion people) primarily depend on medicines derived from animals and plants. For instance, in traditional Chinese medicine, over 1500 animal species have been documented for their medicinal applications[42]. The use of animal-derived products raises a number of issues, from hygiene to ethical ones. Certainly, the utilization of products that do not entail the sacrifice of animals is more ethically acceptable and thus preferable. Deer antlers, which are naturally shed by the animal each year, align with this expectation.

Deer antlers stand as the sole mammalian organ known to fully regenerate naturally once lost. In male deer, the presence of elevated testosterone levels in the bloodstream triggers the onset of antler formation during the second year of life. Antlers emerge as extensions from robust bony projections known as pedicles. Then follows a shift from pedicle to initial antler formation, and growth becomes evident as the integument covering the outgrowth transforms from regular skin (pedicle) to a distinct type of pelage, known as velvet. Due to a rise in circulating testosterone levels, antler growth stops, leading to a complete mineralization of antler bone, and to the shedding of the velvet covering. Following the season of pairings, when testosterone levels fall below this threshold, antlers are dropped (a process known as “antler casting”). In the following years, cycles of periodic regeneration and shedding of a new set of antlers from the pedicles begin[43].

Pedicles and initial antlers originate from a specialized periosteum, known as the antlerogenic periosteum (AP), which, if removed, prevents the formation of the stages. Later, annual antler regeneration is entirely reliant on the presence of cells in pedicle periosteum (PP) tissue. The rapid growth of an antler primarily occurs due to the activity of cells within the proliferation zone, specifically the reserve mesenchyme (RM), and cells within the RM must exhibit significant proliferation potential to sustain such rapid growth[44]. ASCs can be categorized into three types based on their source: Antlerogenic periosteal cells (APCs) originating from the AP, pedicle periosteal cells derived from the PP, and reserve mesenchymal cells (RMCs) originating from the RM[45].

The properties of the cells found in deer antlers, as well as the extracellular structures produced by them, display unique biological characteristics. In traditional Chinese medicine, deer antler extracts are considered to be of central importance in enhancing kidney function, fortifying tendons and bones, and extending longevity, among other purposes. Some scientific works have studied the inhibitory effect of Pilose antler extracts on cancer cells, showing interesting and promising results[46,47].

The cellular products of these cells also possess unique characteristics of clinical interest. Exosomes derived from ASCs are demonstrated to mitigate senescence in human MSCs in vitro[48], and significantly expedite the wound healing process, enhancing its quality, promoting the regeneration of cutaneous appendages (i.e., hair follicles and sebaceous glands), as well as improving the distribution pattern of collagen in the healed skin in a rat model[49]. In another study, it was demonstrated that ASC-derived exosomes were effective in alleviating the symptoms of pulmonary fibrosis in a mouse model system, while also increasing the survival rate of affected mice, in part through modulation of the immune response[50].

Several studies indicate that ASCs exhibit molecular traits akin to pluripotent and multipotent stem cells. This is evidenced, for example, by the fact that c-KIT (stem cell factor receptor) and Sca-1 (stem cell antigen-1), recognized markers of embryonic stem cells and tissue-specific stem cells, respectively, have been detected in over 70% of ASCs[51]. ASCs could be defined as MSCs with embryonic features. Indeed, ASCs express all typical MSC markers, including CD73, CD90, CD105, and STRO-1, along with certain markers associated with embryonic stem cells, such as Tert, Nestin, S100A4, nucleostemin, C-Myc, and Oct-4[52]. Negative expression markers were also assessed by cytofluorometry, in particular CD31, CD45, CD62p, CD133, and HLA-DR, that were absent or poorly expressed[50]. ASCs possess the capability, under particular culture conditions, to differentiate in osteogenic, chondrogenic and adipogenic lineages, as well as into muscle precursor cells and neuron-like cells, if isolated during a specific degree of differentiation[54]. These and other characteristics make ASCs a cell type with high potential for clinical applications.

In the paper commented on here, Wang et al[1] utilized a decellularized xenogeneic ECM derived from ASCs (RMC-ECM) as a substrate for MSC growth, providing evidence that supports the validity of this approach. Fascinatingly, RMC-ECM shows different regenerative properties when produced from cells in quiescent (APCs) or active (RMCs) state. Moreover, when transplanted in rats in order to repair osteochondral defects, ECM derived from deer stem cells, therefore of xenogeneic origin, performed better than allogeneic ECM produced by rat MSCs. The underlying reasons for this difference are still unclear, but some hypotheses can be discussed.

In vitro studies on antlers have effectively isolated and cultured unique ASCs discovered in the AP, PP, and RM, which play a pivotal role in tissue regeneration. Additionally, histological and morphological analyses have revealed the existence of various tissue types within the growth region of the antler[55]. The mechanism of antler regeneration has been extensively investigated, and it is believed that beyond a rise in systemic testosterone levels, a series of other signals from different hormones also intervene, i.e., estrogen, vitamin D, thyroid hormones, and cortisol[56], triggering the histogenesis of the pedicle and serving as the activation signal for the APCs located in the AP. The results shown in the annotated study demonstrate how RMC-ECM could replicate the proliferative effects, naturally shown in deer, in the restoration of osteochondral defects in other animal species by utilizing implantation of cell-free RMC-ECM sheets.

CONCLUSION

Many papers produced in recent years have sought to identify sources of efficient, and possibly cell-free, scaffolds that show high efficiencies and low costs, and that are worthy of exploration for potential applications in human health care. Evaluations of less traditional animal sources, such as deer, compared to those already widely studied such as pigs, may be of great interest for future clinical use.

Another consideration regarding this article, which from our perspective is even more fascinating, is the apparent confirmation that the data reported further supports the idea that the traits of an individual cell, the substances it generates, and the tissue it constitutes can evoke similar phenomena. This suggests a notion of ‘collective memory’ that permeates the entirety of an organism. Our perspective on this topic is that cells, along with all their cellular derivatives, inherently carry information about their own health status and have the capacity to transmit this information. Our group also studied the effects of an animal preparation on cancer human cells, in particular we demonstrated that extracellular vesicles derived from farm animal food derivatives (specifically pigs) can modulate human hepatic cell metabolism, thereby enhancing cell survival even in damaged contexts[57].

It almost seems as if there is a biological will to communicate one’s well-being (as well as any discomfort) to all that surrounds living organisms. This process is undoubtedly mediated by chemical factors, but they alone are not adequate to explain all aspects pertaining to this type of information transmission. There’s a memory that transcends chemistry’s boundaries, a phenomenon we can only define through mechanobiology. A relatively recent concept in the field is that of mechanical memory, which pertains to the enduring effects of mechanical stimuli long after their removal[58]. This memory that crosses the species boundary makes studies similar to the one commented on in the present editorial interesting, paving the way to a new vision of cellular biology where it appears to us that a greater harmony governed by more overarching natural laws may prevail.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Corresponding Author’s Membership in Professional Societies: American Society for Biochemistry and Molecular Biology, 19046.

Specialty type: Cell and tissue engineering

Country of origin: Italy

Peer-review report’s classification

Scientific Quality: Grade A

Novelty: Grade A

Creativity or Innovation: Grade A

Scientific Significance: Grade A

P-Reviewer: Zhang W, China S-Editor: Wang JJ L-Editor: A P-Editor: Che XX

References
1.  Wang YS, Chu WH, Zhai JJ, Wang WY, He ZM, Zhao QM, Li CY. High quality repair of osteochondral defects in rats using the extracellular matrix of antler stem cells. World J Stem Cells. 2024;16:176-190.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (1)]
2.  Ayala-Cuellar AP, Kang JH, Jeung EB, Choi KC. Roles of Mesenchymal Stem Cells in Tissue Regeneration and Immunomodulation. Biomol Ther (Seoul). 2019;27:25-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 82]  [Article Influence: 16.4]  [Reference Citation Analysis (0)]
3.  Huerta CT, Ortiz YY, Liu ZJ, Velazquez OC. Methods and Limitations of Augmenting Mesenchymal Stem Cells for Therapeutic Applications. Adv Wound Care (New Rochelle). 2023;12:467-481.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
4.  Noronha NC, Mizukami A, Caliári-Oliveira C, Cominal JG, Rocha JLM, Covas DT, Swiech K, Malmegrim KCR. Priming approaches to improve the efficacy of mesenchymal stromal cell-based therapies. Stem Cell Res Ther. 2019;10:131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 211]  [Cited by in F6Publishing: 313]  [Article Influence: 62.6]  [Reference Citation Analysis (0)]
5.  Ghasemi-Mobarakeh L, Prabhakaran MP, Tian L, Shamirzaei-Jeshvaghani E, Dehghani L, Ramakrishna S. Structural properties of scaffolds: Crucial parameters towards stem cells differentiation. World J Stem Cells. 2015;7:728-744.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 126]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
6.  Weißenbruch K, Lemma ED, Hippler M, Bastmeyer M. Micro-scaffolds as synthetic cell niches: recent advances and challenges. Curr Opin Biotechnol. 2022;73:290-299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
7.  Wan PX, Wang BW, Wang ZC. Importance of the stem cell microenvironment for ophthalmological cell-based therapy. World J Stem Cells. 2015;7:448-460.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 21]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
8.  Zhao X, Li Q, Guo Z, Li Z. Constructing a cell microenvironment with biomaterial scaffolds for stem cell therapy. Stem Cell Res Ther. 2021;12:583.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 18]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
9.  Reilly GC, Engler AJ. Intrinsic extracellular matrix properties regulate stem cell differentiation. J Biomech. 2010;43:55-62.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 583]  [Cited by in F6Publishing: 526]  [Article Influence: 35.1]  [Reference Citation Analysis (0)]
10.  Osório LA, Silva E, Mackay RE. A Review of Biomaterials and Scaffold Fabrication for Organ-on-a-Chip (OOAC) Systems. Bioengineering (Basel). 2021;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 31]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
11.  Zhan X. Effect of matrix stiffness and adhesion ligand density on chondrogenic differentiation of mesenchymal stem cells. J Biomed Mater Res A. 2020;108:675-683.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 29]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
12.  Zhang J, Wehrle E, Adamek P, Paul GR, Qin XH, Rubert M, Müller R. Optimization of mechanical stiffness and cell density of 3D bioprinted cell-laden scaffolds improves extracellular matrix mineralization and cellular organization for bone tissue engineering. Acta Biomater. 2020;114:307-322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 63]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
13.  El-Rashidy AA, El Moshy S, Radwan IA, Rady D, Abbass MMS, Dörfer CE, Fawzy El-Sayed KM. Effect of Polymeric Matrix Stiffness on Osteogenic Differentiation of Mesenchymal Stem/Progenitor Cells: Concise Review. Polymers (Basel). 2021;13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 17]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
14.  Matsiko A, Gleeson JP, O'Brien FJ. Scaffold mean pore size influences mesenchymal stem cell chondrogenic differentiation and matrix deposition. Tissue Eng Part A. 2015;21:486-497.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 158]  [Cited by in F6Publishing: 163]  [Article Influence: 18.1]  [Reference Citation Analysis (0)]
15.  Wang Y, Kim UJ, Blasioli DJ, Kim HJ, Kaplan DL. In vitro cartilage tissue engineering with 3D porous aqueous-derived silk scaffolds and mesenchymal stem cells. Biomaterials. 2005;26:7082-7094.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 343]  [Cited by in F6Publishing: 353]  [Article Influence: 18.6]  [Reference Citation Analysis (0)]
16.  Midha S, Jain KG, Bhaskar N, Kaur A, Rawat S, Giri S, Basu B, Mohanty S. Tissue-specific mesenchymal stem cell-dependent osteogenesis in highly porous chitosan-based bone analogs. Stem Cells Transl Med. 2021;10:303-319.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
17.  Spreda M, Hauptmann N, Lehner V, Biehl C, Liefeith K, Lips KS. Porous 3D Scaffolds Enhance MSC Vitality and Reduce Osteoclast Activity. Molecules. 2021;26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
18.  Xiao L, Sun Y, Liao L, Su X. Response of mesenchymal stem cells to surface topography of scaffolds and the underlying mechanisms. J Mater Chem B. 2023;11:2550-2567.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 4]  [Reference Citation Analysis (0)]
19.  Lužanin O, Gudurić V, Bernhardt A, Movrin D, Damjanović-Vasilić L, Terek P, Ostojić G, Stankovski S. Impact of In-Process Crystallinity of Biodegradable Scaffolds Fabricated by Material Extrusion on the Micro- and Nanosurface Topography, Viability, Proliferation, and Differentiation of Human Mesenchymal Stromal Cells. Polymers (Basel). 2023;15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
20.  Kao TW, Chiou A, Lin KH, Liu YS, Lee OK. Alteration of 3D Matrix Stiffness Regulates Viscoelasticity of Human Mesenchymal Stem Cells. Int J Mol Sci. 2021;22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
21.  Whitehead J, Griffin KH, Gionet-Gonzales M, Vorwald CE, Cinque SE, Leach JK. Hydrogel mechanics are a key driver of bone formation by mesenchymal stromal cell spheroids. Biomaterials. 2021;269:120607.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 40]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
22.  Xing Z, Cai J, Sun Y, Cao M, Li Y, Xue Y, Finne-Wistrand A, Kamal M. Altered Surface Hydrophilicity on Copolymer Scaffolds Stimulate the Osteogenic Differentiation of Human Mesenchymal Stem Cells. Polymers (Basel). 2020;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
23.  Yang J, Xiao Y, Tang Z, Luo Z, Li D, Wang Q, Zhang X. The negatively charged microenvironment of collagen hydrogels regulates the chondrogenic differentiation of bone marrow mesenchymal stem cells in vitro and in vivo. J Mater Chem B. 2020;8:4680-4693.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
24.  Kharbikar BN, Mohindra P, Desai TA. Biomaterials to enhance stem cell transplantation. Cell Stem Cell. 2022;29:692-721.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 30]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
25.  Neishabouri A, Soltani Khaboushan A, Daghigh F, Kajbafzadeh AM, Majidi Zolbin M. Decellularization in Tissue Engineering and Regenerative Medicine: Evaluation, Modification, and Application Methods. Front Bioeng Biotechnol. 2022;10:805299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 47]  [Article Influence: 23.5]  [Reference Citation Analysis (0)]
26.  Poel WE. Preparation of Acellular Homogenates From Muscle Samples. Science. 1948;108:390-391.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 20]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
27.  Zhang X, Chen X, Hong H, Hu R, Liu J, Liu C. Decellularized extracellular matrix scaffolds: Recent trends and emerging strategies in tissue engineering. Bioact Mater. 2022;10:15-31.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 216]  [Article Influence: 108.0]  [Reference Citation Analysis (0)]
28.  Rana D, Zreiqat H, Benkirane-Jessel N, Ramakrishna S, Ramalingam M. Development of decellularized scaffolds for stem cell-driven tissue engineering. J Tissue Eng Regen Med. 2017;11:942-965.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 119]  [Cited by in F6Publishing: 135]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
29.  O'Brien MF, Goldstein S, Walsh S, Black KS, Elkins R, Clarke D. The SynerGraft valve: a new acellular (nonglutaraldehyde-fixed) tissue heart valve for autologous recellularization first experimental studies before clinical implantation. Semin Thorac Cardiovasc Surg. 1999;11:194-200.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Kafili G, Kabir H, Jalali Kandeloos A, Golafshan E, Ghasemi S, Mashayekhan S, Taebnia N. Recent advances in soluble decellularized extracellular matrix for heart tissue engineering and organ modeling. J Biomater Appl. 2023;38:577-604.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Reference Citation Analysis (0)]
31.  Wang M, Wu Y, Li G, Lin Q, Zhang W, Liu H, Su J. Articular cartilage repair biomaterials: strategies and applications. Mater Today Bio. 2024;24:100948.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
32.  Discher DE, Mooney DJ, Zandstra PW. Growth factors, matrices, and forces combine and control stem cells. Science. 2009;324:1673-1677.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2129]  [Cited by in F6Publishing: 1819]  [Article Influence: 121.3]  [Reference Citation Analysis (0)]
33.  Marinkovic M, Block TJ, Rakian R, Li Q, Wang E, Reilly MA, Dean DD, Chen XD. One size does not fit all: developing a cell-specific niche for in vitro study of cell behavior. Matrix Biol. 2016;52-54:426-441.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 62]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
34.  Urciuolo A, Urbani L, Perin S, Maghsoudlou P, Scottoni F, Gjinovci A, Collins-Hooper H, Loukogeorgakis S, Tyraskis A, Torelli S, Germinario E, Fallas MEA, Julia-Vilella C, Eaton S, Blaauw B, Patel K, De Coppi P. Decellularised skeletal muscles allow functional muscle regeneration by promoting host cell migration. Sci Rep. 2018;8:8398.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 48]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
35.  Shimoda H, Yagi H, Higashi H, Tajima K, Kuroda K, Abe Y, Kitago M, Shinoda M, Kitagawa Y. Decellularized liver scaffolds promote liver regeneration after partial hepatectomy. Sci Rep. 2019;9:12543.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 42]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
36.  Stocco E, Barbon S, Mammana M, Zambello G, Contran M, Parnigotto PP, Macchi V, Conconi MT, Rea F, De Caro R, Porzionato A. Preclinical and clinical orthotopic transplantation of decellularized/engineered tracheal scaffolds: A systematic literature review. J Tissue Eng. 2023;14:20417314231151826.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 8]  [Reference Citation Analysis (0)]
37.  Massaro MS, Pálek R, Rosendorf J, Červenková L, Liška V, Moulisová V. Decellularized xenogeneic scaffolds in transplantation and tissue engineering: Immunogenicity versus positive cell stimulation. Mater Sci Eng C Mater Biol Appl. 2021;127:112203.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 10]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
38.  Eble JA, Niland S. The extracellular matrix in tumor progression and metastasis. Clin Exp Metastasis. 2019;36:171-198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in F6Publishing: 312]  [Article Influence: 62.4]  [Reference Citation Analysis (0)]
39.  Hoshiba T, Tanaka M. Breast cancer cell behaviors on staged tumorigenesis-mimicking matrices derived from tumor cells at various malignant stages. Biochem Biophys Res Commun. 2013;439:291-296.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 28]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
40.  Dunne LW, Huang Z, Meng W, Fan X, Zhang N, Zhang Q, An Z. Human decellularized adipose tissue scaffold as a model for breast cancer cell growth and drug treatments. Biomaterials. 2014;35:4940-4949.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 104]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
41.  Li C, Li Y, Wang W, Scimeca M, Melino G, Du R, Shi Y. Deer antlers: the fastest growing tissue with least cancer occurrence. Cell Death Differ. 2023;30:2452-2461.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
42.  Alves RR, Rosa IL. Why study the use of animal products in traditional medicines? J Ethnobiol Ethnomed. 2005;1:5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 53]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
43.  Kierdorf U, Kierdorf H. Deer antlers - a model of mammalian appendage regeneration: an extensive review. Gerontology. 2011;57:53-65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 60]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
44.  Zhang W, Ke CH, Guo HH, Xiao L. Antler stem cells and their potential in wound healing and bone regeneration. World J Stem Cells. 2021;13:1049-1057.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 2.0]  [Reference Citation Analysis (1)]
45.  Liu Q, Li J, Chang J, Guo Y, Wen D. The characteristics and medical applications of antler stem cells. Stem Cell Res Ther. 2023;14:225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
46.  Li M, Li Q, Dong H, Zhao S, Ning J, Bai X, Yue X, Xie A. Pilose antler polypeptides enhance chemotherapy effects in triple-negative breast cancer by activating the adaptive immune system. Int J Biol Macromol. 2022;222:2628-2638.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
47.  Liu G, Ma C, Wang P, Zhang P, Qu X, Liu S, Zhai Z, Yu D, Gao J, Liang J, Dai W, Zhou L, Xia M, Yang H. Pilose antler peptide potentiates osteoblast differentiation and inhibits osteoclastogenesis via manipulating the NF-κB pathway. Biochem Biophys Res Commun. 2017;491:388-395.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 22]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
48.  Lei J, Jiang X, Li W, Ren J, Wang D, Ji Z, Wu Z, Cheng F, Cai Y, Yu ZR, Belmonte JCI, Li C, Liu GH, Zhang W, Qu J, Wang S. Exosomes from antler stem cells alleviate mesenchymal stem cell senescence and osteoarthritis. Protein Cell. 2022;13:220-226.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
49.  Zhang G, Wang D, Ren J, Li J, Guo Q, Shi L, Li C. Antler stem cell-derived exosomes promote regenerative wound healing via fibroblast-to-myofibroblast transition inhibition. J Biol Eng. 2023;17:67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
50.  Zhang G, Shi L, Li J, Wang S, Ren J, Wang D, Hu P, Wang Y, Li C. Antler stem cell exosomes alleviate pulmonary fibrosis via inhibiting recruitment of monocyte macrophage, rather than polarization of M2 macrophages in mice. Cell Death Discov. 2023;9:359.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
51.  Daley EL, Alford AI, Miller JD, Goldstein SA. Phenotypic differences in white-tailed deer antlerogenic progenitor cells and marrow-derived mesenchymal stromal cells. Tissue Eng Part A. 2014;20:1416-1425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
52.  Wang D, Berg D, Ba H, Sun H, Wang Z, Li C. Deer antler stem cells are a novel type of cells that sustain full regeneration of a mammalian organ-deer antler. Cell Death Dis. 2019;10:443.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 30]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
53.  Seo MS, Park SB, Choi SW, Kim JJ, Kim HS, Kang KS. Isolation and characterization of antler-derived multipotent stem cells. Cell Transplant. 2014;23:831-843.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 15]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
54.  Li C, Yang F, Sheppard A. Adult stem cells and mammalian epimorphic regeneration-insights from studying annual renewal of deer antlers. Curr Stem Cell Res Ther. 2009;4:237-251.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 69]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
55.  Li C, Clark DE, Lord EA, Stanton JA, Suttie JM. Sampling technique to discriminate the different tissue layers of growing antler tips for gene discovery. Anat Rec. 2002;268:125-130.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 77]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
56.  Feleke M, Bennett S, Chen J, Hu X, Williams D, Xu J. New physiological insights into the phenomena of deer antler: A unique model for skeletal tissue regeneration. J Orthop Translat. 2021;27:57-66.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 5]  [Article Influence: 1.3]  [Reference Citation Analysis (1)]
57.  Tassinari R, Cavallini C, Olivi E, Taglioli V, Zannini C, Ferroni O, Ventura C. Protective effects of exosomes derived from lyophilized porcine liver against acetaminophen damage on HepG2 cells. BMC Complement Med Ther. 2021;21:299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
58.  Lele TP, Brock A, Peyton SR. Emerging Concepts and Tools in Cell Mechanomemory. Ann Biomed Eng. 2020;48:2103-2112.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 1]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]