1
|
Hodgson-Garms M, Moore MJ, Martino MM, Kelly K, Frith JE. Proteomic profiling of iPSC and tissue-derived MSC secretomes reveal a global signature of inflammatory licensing. NPJ Regen Med 2025; 10:7. [PMID: 39905050 PMCID: PMC11794695 DOI: 10.1038/s41536-024-00382-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 12/03/2024] [Indexed: 02/06/2025] Open
Abstract
Much of the therapeutic potential of mesenchymal stromal cells (MSCs) is underpinned by their secretome which varies significantly with source, donor and microenvironmental cues. Understanding these differences is essential to define the mechanisms of MSC-based tissue repair and optimise cell therapies. This study analysed the secretomes of bone-marrow (BM.MSCs), umbilical-cord (UC.MSCs), adipose-tissue (AT.MSCs) and clinical/commercial-grade induced pluripotent stem cell-derived MSCs (iMSCs), under resting and inflammatory licenced conditions. iMSCs recapitulated the inflammatory licensing process, validating their comparability to tissue-derived MSCs. Overall, resting secretomes were defined by extracellular matrix (ECM) and pro-regenerative proteins, while licensed secretomes were enriched in chemotactic and immunomodulatory proteins. iMSC and UC.MSC secretomes contained proteins indicating proliferative potential and telomere maintenance, whereas adult tissue-derived secretomes contained fibrotic and ECM-related proteins. The data and findings from this study will inform the optimum MSC source for particular applications and underpin further development of MSC therapies.
Collapse
Affiliation(s)
- Margeaux Hodgson-Garms
- Department of Materials Science and Engineering, Monash University, Melbourne, VIC, Australia.
- Cynata Therapeutics, Melbourne, VIC, Australia.
| | - Matthew J Moore
- Department of Materials Science and Engineering, Monash University, Melbourne, VIC, Australia
| | - Mikaël M Martino
- Australian Regenerative Medicine Institute, Melbourne, VIC, Australia
- Victorian Heart Institute, Monash University, Melbourne, VIC, Australia
| | | | - Jessica E Frith
- Department of Materials Science and Engineering, Monash University, Melbourne, VIC, Australia.
- Australian Regenerative Medicine Institute, Melbourne, VIC, Australia.
| |
Collapse
|
2
|
Fath MK, Zadian SS, Torbati SMB, Saqagandomabadi V, Afshar OY, Khalilzad M, Abedi S, Moliani A, Barati G. Roles of Mesenchymal Stem Cells in Breast Cancer Therapy: Engineered Stem Cells and Exosomal Cell-Free Based Therapy. Curr Mol Med 2025; 25:431-444. [PMID: 38275063 DOI: 10.2174/0115665240274818231207054037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/03/2023] [Accepted: 11/07/2023] [Indexed: 01/27/2024]
Abstract
Breast cancer has a high prevalence among women, with a high mortality rate. The number of people who suffer from breast cancer disease is increasing, whereas metastatic cancers are mostly incurable, and existing therapies have unfavorable side effects. For an extended duration, scientists have dedicated their efforts to exploring the potential of mesenchymal stem cells (MSCs) for the treatment of metastatic cancers, including breast cancer. MSCs could be genetically engineered to boost their anticancer potency. Furthermore, MSCs can transport oncolytic viruses, suicide genes, and anticancer medicines to tumors. Extracellular vesicles (EVs) are MSC products that have attracted scientist's attention as a cell-free treatment. This study narratively reviews the current state of knowledge on engineered MSCs and their EVs as promising treatments for breast cancer.
Collapse
Affiliation(s)
- Mohsen Karami Fath
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Seyed Sajjad Zadian
- Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Vahid Saqagandomabadi
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | | | - Mohammad Khalilzad
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Abedi
- Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Afshin Moliani
- Isfahan Medical Students Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | | |
Collapse
|
3
|
Baran Z, Çetinkaya M, Baran Y. Mesenchymal Stem Cells in Cancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1474:149-177. [PMID: 39470980 DOI: 10.1007/5584_2024_824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2024]
Abstract
The mesenchymal stem/stromal cells (MSCs) are multipotent cells that were initially discovered in the bone marrow in the late 1960s but have so far been discovered in almost all tissues of the body. The multipotent property of MSCs enables them to differentiate into various cell types and lineages, such as adipocytes, chondrocytes, and osteocytes. The immunomodulation capacity and tumor-targeting features of MSCs made their use crucial for cell-based therapies in cancer treatment, yet limited advancement could be observed in translational medicine prospects due to the need for more information regarding the controversial roles of MSCs in crosstalk tumors. In this review, we discuss the therapeutic potential of MSCs, the controversial roles played by MSCs in cancer progression, and the anticancer therapeutic strategies that are in association with MSCs. Finally, the clinical trials designed for the direct use of MSCs for cancer therapy or for their use in decreasing the side effects of other cancer therapies are also mentioned in this review to evaluate the current status of MSC-based cancer therapies.
Collapse
Affiliation(s)
- Züleyha Baran
- Laboratory of Molecular Pharmacology, Department of Pharmacology, Anadolu University, Eskişehir, Turkey
| | - Melisa Çetinkaya
- Laboratory of Cancer Genetics, Department of Molecular Biology and Genetics, İzmir Institute of Technology, İzmir, Turkey
| | - Yusuf Baran
- Laboratory of Cancer Genetics, Department of Molecular Biology and Genetics, İzmir Institute of Technology, İzmir, Turkey.
| |
Collapse
|
4
|
Teng FS, de Faria Lainetti P, Simão Franzoni M, Fernando Leis Filho A, de Oliveira Massoco Salles Gomes C, Laufer-Amorim R, Martins Amorim R, Fonseca-Alves CE. Canine Adipose-Derived Mesenchymal Stromal Cells Reduce Cell Viability and Migration of Metastatic Canine Oral Melanoma Cell Lines In Vitro. Vet Sci 2024; 11:636. [PMID: 39728976 PMCID: PMC11680336 DOI: 10.3390/vetsci11120636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 11/28/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024] Open
Abstract
Canine oral melanoma (COM) is a promising target for immunomodulatory therapies aimed at enhancing the immune system's antitumor response. Given that adipose-derived mesenchymal stem cells (Ad-MSCs) possess immunomodulatory properties through cytokine release, we hypothesized that co-culturing Ad-MSCs and canine peripheral blood mononuclear cells (PBMCs) could stimulate interleukin (IL) production against melanoma cell lines (MCCLs) and help identify therapeutic targets. This study evaluated IL-2, IL-8, and IL-12 expressions in co-culture with MCCL, Ad-MSCs, and PBMCs and assessed the relationship between gene expression, cell viability, and migration. Using four experimental groups in a Transwell insert system to separate cell types, we found that Ad-MSCs can reduce MCCL migration and viability, though the effect may vary depending on each cell line's susceptibility. Furthermore, Ad-MSCs modified IL expression profiles in co-cultured cells. Our findings suggest that Ad-MSCs could have therapeutic potential for COM by inhibiting cell migration and reducing viability. However, deeper insights into Ad-MSC interactions with the tumor microenvironment and melanoma-specific factors will be essential to optimize therapeutic efficacy.
Collapse
Affiliation(s)
- Fwu Shing Teng
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu 18618-681, Brazil; (F.S.T.); (P.d.F.L.); (M.S.F.); (A.F.L.F.)
| | - Patricia de Faria Lainetti
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu 18618-681, Brazil; (F.S.T.); (P.d.F.L.); (M.S.F.); (A.F.L.F.)
| | - Mayara Simão Franzoni
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu 18618-681, Brazil; (F.S.T.); (P.d.F.L.); (M.S.F.); (A.F.L.F.)
| | - Antonio Fernando Leis Filho
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu 18618-681, Brazil; (F.S.T.); (P.d.F.L.); (M.S.F.); (A.F.L.F.)
| | | | - Renée Laufer-Amorim
- Department of Veterinary Clinic, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu 18618-681, Brazil; (R.L.-A.); (R.M.A.)
| | - Rogério Martins Amorim
- Department of Veterinary Clinic, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu 18618-681, Brazil; (R.L.-A.); (R.M.A.)
| | - Carlos Eduardo Fonseca-Alves
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu 18618-681, Brazil; (F.S.T.); (P.d.F.L.); (M.S.F.); (A.F.L.F.)
- Institute of Veterinary Oncology (IOVET), Sao Paulo 05027-020, Brazil
- Vet Precision Laboratory, Botucatu 18610-034, Brazil
| |
Collapse
|
5
|
Zhang WJ, Chen D. Mesenchymal stem cell transplantation plays a role in relieving cancer pain. Front Pharmacol 2024; 15:1483716. [PMID: 39679363 PMCID: PMC11637888 DOI: 10.3389/fphar.2024.1483716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024] Open
Abstract
Tumors can invade, compress, and damage nerves, leading to persistent pain and seriously affecting the quality of life of patients. However, their treatment is challenging. Sensitization of peripheral receptors, abnormal activity of primary sensory neurons, activation of glial cells, enhanced inflammatory responses, and sensory information transmission contribute towards cancer pain. Therefore, considerable attention has been paid to exploring prospective methods to inhibit the occurrence of these factors and relieve cancer pain. Studies on different types of pains have revealed that the transplantation of functionally active cells into the host has the pharmacological effect of producing analgesia. Mesenchymal stem cells (MSCs) can act as small active pumps to reduce the expression of pain-related molecules and produce analgesic effects. Moreover, MSCs can establish complex communication networks with non-tumor and cancer cells in the microenvironment, interact with each other, and can be used as destinations for inflammation and tumor sites, affecting their potential for invasion and metastasis. This emphasizes the key role of MSCs in cancer and pain management. The pain relief mechanisms of MSCs include neuronutrition, neural protection, neural network reconstruction, immune regulation, and improvement of the inflammatory microenvironment around the nerve injury. All of these are beneficial for the recovery of injured or stimulated nerves and the reconstruction of neural function, and play a role in relieving pain. The pain treatment strategy of cell transplantation is to repair injured nerves and produce analgesic pharmacological properties that are different from those of painkillers and other physiotherapies. Although the therapeutic role of MSCs in cancer and pain is in its early stages, the therapeutic value of MSCs for cancer pain has great prospects. Therefore, in this study, we explored the possible mechanism between MSCs and cancer pain, the potential therapeutic role of therapeutic cells in cancer pain, and some problems and challenges.
Collapse
Affiliation(s)
- Wen-Jun Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Nanchang University, Jiangxi Medical college, Nanchang, China
| | - Dingyi Chen
- Emergency department, The Second Affiliated Hospital, Nanchang University, Jiangxi Medical college, Nanchang, China
| |
Collapse
|
6
|
Li Y, Jin M, Guo D, Shen S, Lu K, Pan R, Sun L, Zhang H, Shao J, Pan G. Unveiling the immunogenicity of allogeneic mesenchymal stromal cells: Challenges and strategies for enhanced therapeutic efficacy. Biomed Pharmacother 2024; 180:117537. [PMID: 39405918 DOI: 10.1016/j.biopha.2024.117537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 11/14/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) exhibit significant potential in the context of cell therapy because of their capacity to perform a range of interconnected functions in damaged tissues, including immune modulation, hematopoietic support, and tissue regeneration. MSCs are hypoimmunogenic because of their diminished expression of major histocompatibility molecules, absence of costimulatory molecules, and presence of coinhibitory molecules. While autologous MSCs reduce the risk of rejection and infection, variability in cell numbers and proliferation limits their potential applications. Conversely, allogeneic MSCs (allo-MSCs) possess broad clinical applications unconstrained by donor physiology. Nonetheless, preclinical and clinical investigations highlight that transplanted allo-MSCs are subject to immune attack from recipients. These cells exhibit anti-inflammatory and proinflammatory phenotypes contingent on the microenvironment. Notably, the proinflammatory phenotype features enhanced immunogenicity and diminished immunosuppression, potentially triggering allogeneic immune reactions that impede long-term clinical efficacy. Consequently, preserving the low immunogenicity of allo-MSCs in vivo and mitigating immune rejection in diverse microenvironments represent crucial challenges for the widespread clinical application of MSCs. In this review, we elucidate the immune regulation of allo-MSCs, specifically focusing on two distinct subgroups, MSC1 and MSC2, that exhibit varying polarization states and immunogenicity. We discuss the factors and underlying mechanisms that induce MSC immunogenicity and polarization, highlighting the crucial role of major histocompatibility complex class I/II molecules in rejection post-transplantation. Additionally, we summarize the immunogenic regulatory targets and applications of allo-MSCs and outline strategies to address challenges in this promising field, aiming to enhance allo-MSC therapeutic efficacy for patients.
Collapse
Affiliation(s)
- Yuanhui Li
- Department of Oncological Surgery, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China
| | - Mengting Jin
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Dongyang Guo
- Hangzhou City University, School of Medicine, 50 Huzhou Street, Hangzhou, China
| | - Shuang Shen
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Kaining Lu
- Breast Disease Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ruolang Pan
- Key Laboratory of Cell-Based Drug and Applied Technology Development in Zhejiang Province, Hangzhou, China
| | - Li Sun
- Department of Oncological Surgery, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China
| | - Hongchen Zhang
- Department of Gatroenterology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, No. 261 HuanSha Road, Hangzhou, China.
| | - Jianzhong Shao
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang University, Hangzhou, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| | - Gang Pan
- Department of Oncological Surgery, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China.
| |
Collapse
|
7
|
Oishi T, Koizumi S, Kurozumi K. Mesenchymal stem cells as therapeutic vehicles for glioma. Cancer Gene Ther 2024; 31:1306-1314. [PMID: 38654128 DOI: 10.1038/s41417-024-00775-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024]
Abstract
Glioma is a disease with a poor prognosis despite the availability of multimodality treatments, and the development of novel therapies is urgently needed. Challenges in glioma treatment include the difficulty for drugs to cross the blood-brain barrier when administered systemically and poor drug diffusion when administered locally. Mesenchymal stem cells exhibit advantages for glioma therapy because of their ability to pass through the blood-brain barrier and migrate to tumor cells and their tolerance to the immune system. Therefore, mesenchymal stem cells have been explored as vehicles for various therapeutic agents for glioma treatment. Mesenchymal stem cells loaded with chemotherapeutic drugs show improved penetration and tumor accumulation. For gene therapy, mesenchymal stem cells can be used as vehicles for suicide genes, the so-called gene-directed enzyme prodrug therapy. Mesenchymal stem cell-based oncolytic viral therapies have been attempted in recent years to enhance the efficacy of infection against the tumor, viral replication, and distribution of viral particles. Many uncertainties remain regarding the function and behavior of mesenchymal stem cells in gliomas. However, strategies to increase mesenchymal stem cell migration to gliomas may improve the delivery of therapeutic agents and enhance their anti-tumor effects, representing promising potential for patient treatment.
Collapse
Affiliation(s)
- Tomoya Oishi
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Shinichiro Koizumi
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuhiko Kurozumi
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| |
Collapse
|
8
|
Garza Treviño EN, Quiroz Reyes AG, Delgado Gonzalez P, Rojas Murillo JA, Islas JF, Alonso SS, Gonzalez Villarreal CA. Applications of Modified Mesenchymal Stem Cells as Targeted Systems against Tumor Cells. Int J Mol Sci 2024; 25:7791. [PMID: 39063032 PMCID: PMC11276748 DOI: 10.3390/ijms25147791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
Combined gene and cell therapy are promising strategies for cancer treatment. Given the complexity of cancer, several approaches are actively studied to fight this disease. Using mesenchymal stem cells (MSCs) has demonstrated dual antitumor and protumor effects as they exert massive immune/regulatory effects on the tissue microenvironment. MSCs have been widely investigated to exploit their antitumor target delivery system. They can be genetically modified to overexpress genes and selectively or more efficiently eliminate tumor cells. Current approaches tend to produce more effective and safer therapies using MSCs or derivatives; however, the effect achieved by engineered MSCs in solid tumors is still limited and depends on several factors such as the cell source, transgene, and tumor target. This review describes the progress of gene and cell therapy focused on MSCs as a cornerstone against solid tumors, addressing the different MSC-engineering methods that have been approached over decades of research. Furthermore, we summarize the main objectives of engineered MSCs against the most common cancers and discuss the challenges, limitations, risks, and advantages of targeted treatments combined with conventional ones.
Collapse
Affiliation(s)
- Elsa N. Garza Treviño
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (E.N.G.T.); (A.G.Q.R.); (P.D.G.); (J.A.R.M.); (J.F.I.)
| | - Adriana G. Quiroz Reyes
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (E.N.G.T.); (A.G.Q.R.); (P.D.G.); (J.A.R.M.); (J.F.I.)
| | - Paulina Delgado Gonzalez
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (E.N.G.T.); (A.G.Q.R.); (P.D.G.); (J.A.R.M.); (J.F.I.)
| | - Juan Antonio Rojas Murillo
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (E.N.G.T.); (A.G.Q.R.); (P.D.G.); (J.A.R.M.); (J.F.I.)
| | - Jose Francisco Islas
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (E.N.G.T.); (A.G.Q.R.); (P.D.G.); (J.A.R.M.); (J.F.I.)
| | - Santiago Saavedra Alonso
- Departamento de Ciencias Básicas, Vicerrectoría de Ciencias de la Salud, Universidad de Monterrey, Ignacio Morones Prieto 4500, Jesus M. Garza, San Pedro Garza García 66238, Nuevo León, Mexico
| | - Carlos A. Gonzalez Villarreal
- Departamento de Ciencias Básicas, Vicerrectoría de Ciencias de la Salud, Universidad de Monterrey, Ignacio Morones Prieto 4500, Jesus M. Garza, San Pedro Garza García 66238, Nuevo León, Mexico
| |
Collapse
|
9
|
Kaçaroğlu D, Yaylacı S, Gurbuz N. Anti-tumorigenic effects of naive and TLR4-primed adipose-derived mesenchymal stem cells on pancreatic ductal adenocarcinoma cells. Cancer Med 2024; 13:e6964. [PMID: 38379331 PMCID: PMC10831913 DOI: 10.1002/cam4.6964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/09/2024] [Accepted: 01/14/2024] [Indexed: 02/22/2024] Open
Abstract
BACKGROUND One of the main reasons for the unsuccessful treatment of pancreatic cancer is the intense desmoplastic pancreatic microenvironment. In the literature, the effects of mesenchymal stem cells (MSCs) and their inflammatory phenotypes on cancer cells have been a subject of controversy. Therefore, it is crucial to elucidate the underlying mechanisms of this interaction, especially in the context of pancreatic cancer. We aimed to investigate the effects of naive, TLR4-activated, and TLR4-inhibited phenotypes of adipose-derived MSCs (ADMSC) on pancreatic ductal cell line (Panc-1). METHODS AND MATERIALS Adipose-derived MSCs were induced into a proinflammatory phenotype using a 0.5 μg/mL dose of TLR4 agonist, while an anti-inflammatory phenotype was generated in ADMSCs using a 25 μg/mL dose of TLR4 antagonist. We observed that the proliferation of Panc-1 cells was inhibited when naive ADMSCs:Panc-1(10:1) and proinflammatory ADMSCs:Panc-1(10:1) were directly cocultured. RESULTS In indirect coculture, both naive and proinflammatory ADMSCs exhibited a significant 10-fold increase in their inhibitory effect on the proliferation and colony forming capacity of Panc-1 cells, with the added benefit of inducing apoptosis. In our study, both naive and proinflammatory ADMSCs were found to regulate the expression of genes associated with metastasis (MMP2, KDR, MMP9, TIMP1, IGF2R, and COL1A1) and EMT (CDH1, VIM, ZEB1, and CLDN1) in Panc-1 cells. Remarkably, both naive and proinflammatory ADMSCs demonstrated antitumor effects on Panc-1 cells. However, it was observed that anti-inflammatory ADMSCs showed tumor-promoting effects instead. Furthermore, we observed a reciprocal influence between ADMSCs and Panc-1 cells on each other's proinflammatory cytokine expressions, suggesting a dynamic interplay within the tumor microenvironment. CONCLUSIONS These findings underscore the significance of both the naive state and different inflammatory phenotypes of MSCs in the microenvironment and represent a pivotal step toward the development of novel therapeutic approaches for pancreatic cancer. Understanding the intricate interactions between MSCs and cancer cells may open new avenues for targeted interventions in cancer therapy.
Collapse
Affiliation(s)
- Demet Kaçaroğlu
- Department of Medical Biology, Faculty of MedicineSuleyman Demirel UniversityIspartaTurkey
- Department of Medical Biology, Faculty of MedicineLokman Hekim UniversityAnkaraTurkey
| | - Seher Yaylacı
- Department of Medical Biology, Faculty of MedicineLokman Hekim UniversityAnkaraTurkey
| | - Nilgun Gurbuz
- Department of Medical Biology, Faculty of MedicineSuleyman Demirel UniversityIspartaTurkey
| |
Collapse
|
10
|
Smolinska V, Harsanyi S, Bohac M, Danisovic L. Exploring the Three-Dimensional Frontier: Advancements in MSC Spheroids and Their Implications for Breast Cancer and Personalized Regenerative Therapies. Biomedicines 2023; 12:52. [PMID: 38255159 PMCID: PMC10813175 DOI: 10.3390/biomedicines12010052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 01/24/2024] Open
Abstract
To more accurately replicate the in vivo three-dimensional (3D) mesenchymal stem cell (MSC) niche and enhance cellular phenotypes for superior in vivo treatments, MSC functionalization through in vitro 3D culture approaches has gained attention. The organization of MSCs in 3D spheroids results in altered cell shape, cytoskeleton rearrangement, and polarization. Investigations have revealed that the survival and secretory capability of MSCs are positively impacted by moderate hypoxia within the inner zones of MSC spheroids. The spheroid hypoxic microenvironment enhances the production of angiogenic and anti-apoptotic molecules, including HGF, VEGF, and FGF-2. Furthermore, it upregulates the expression of hypoxia-adaptive molecules such as CXCL12 and HIF-1, inhibiting MSC death. The current review focuses on the latest developments in fundamental and translational research concerning three-dimensional MSC systems. This emphasis extends to the primary benefits and potential applications of MSC spheroids, particularly in the context of breast cancer and customized regenerative therapies.
Collapse
Affiliation(s)
- Veronika Smolinska
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia; (V.S.); (M.B.); (L.D.)
| | - Stefan Harsanyi
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia; (V.S.); (M.B.); (L.D.)
| | - Martin Bohac
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia; (V.S.); (M.B.); (L.D.)
- Regenmed Ltd., Medena 29, 811 02 Bratislava, Slovakia
| | - Lubos Danisovic
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia; (V.S.); (M.B.); (L.D.)
- Regenmed Ltd., Medena 29, 811 02 Bratislava, Slovakia
| |
Collapse
|
11
|
Cassano JM, Leonard BC, Martins BC, Vapniarsky N, Morgan JT, Dow SW, Wotman KL, Pezzanite LM. Preliminary evaluation of safety and migration of immune activated mesenchymal stromal cells administered by subconjunctival injection for equine recurrent uveitis. Front Vet Sci 2023; 10:1293199. [PMID: 38162475 PMCID: PMC10757620 DOI: 10.3389/fvets.2023.1293199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 11/29/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction Equine recurrent uveitis (ERU), an immune mediated disease characterized by repeated episodes of intra-ocular inflammation, affects 25% of horses in the USA and is the most common cause of glaucoma, cataracts, and blindness. Mesenchymal stromal cells (MSCs) have immunomodulatory properties, which are upregulated by preconditioning with toll-like receptor agonists. The objective was to evaluate safety and migration of TLR-3 agonist polyinosinic, polycytidylic acid (pIC)-activated MSCs injected subconjunctivally in healthy horses prior to clinical application in horses with ERU. We hypothesized that activated allogeneic MSCs injected subconjunctivally would not induce ocular or systemic inflammation and would remain in the conjunctiva for >14 days. Methods Bulbar subconjunctiva of two horses was injected with 10 × 106 pIC-activated (10 μg/mL, 2 h) GFP-labeled MSCs from one donor three times at two-week intervals. Vehicle (saline) control was injected in the contralateral conjunctiva. Horses received physical and ophthalmic exams [slit lamp biomicroscopy, rebound tonometry, fundic examination, and semiquantitative preclinical ocular toxicology scoring (SPOTS)] every 1-3 days. Systemic inflammation was assessed via CBC, fibrinogen, and serum amyloid A (SAA). Horses were euthanized 14 days following final injection. Full necropsy and histopathology were performed to examine ocular tissues and 36 systemic organs for MSC presence via IVIS Spectrum. Anti-GFP immunohistochemistry was performed on ocular tissues. Results No change in physical examinations was noted. Bloodwork revealed fibrinogen 100-300 mg/dL (ref 100-400) and SAA 0-25 μg/mL (ref 0-20). Ocular effects of the subjconjucntival injection were similar between MSC and control eyes on SPOTS grading system, with conjunctival hypermia, chemosis and ocular discharge noted bilaterally, which improved without intervention within 14 days. All other ocular parameters were unaffected throughout the study. Necropsy and histopathology revealed no evidence of systemic inflammation. Ocular histopathology was similar between MSC and control eyes. Fluorescent imaging analysis did not locate MSCs. Immunohistochemistry did not identify intact MSCs in the conjunctiva, but GFP-labeled cellular components were present in conjunctival phagocytic cells. Discussion Allogeneic pIC-activated conjunctival MSC injections were well tolerated. GFP-labeled tracking identified MSC components phagocytosed by immune cells subconjunctivally. This preliminary safety and tracking information is critical towards advancing immune conditioned cellular therapies to clinical trials in horses.
Collapse
Affiliation(s)
- Jennifer M. Cassano
- Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Brian C. Leonard
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
- Department of Ophthalmology & Vision Science, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Bianca C. Martins
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Natalia Vapniarsky
- Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Joshua T. Morgan
- Department of Bioengineering, University of California, Riverside, Riverside, CA, United States
| | - Steven W. Dow
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Kathryn L. Wotman
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Lynn M. Pezzanite
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
12
|
Ramírez Idarraga JA, Restrepo Múnera LM. Mesenchymal Stem Cells: Their Role in the Tumor Microenvironment. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:681-691. [PMID: 37276173 DOI: 10.1089/ten.teb.2023.0048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Mesenchymal stem cells (MSCs) have been seen for years as great candidates for treating different diseases and an alternative to embryonic stem cells due to their differentiation capacity in vitro. More recent research has focused on their ability to modulate the immune response and regeneration at sites associated with inflammation, activities attributable to the release of trophic factors into the extracellular medium, a set of components known as the secretome. It has been possible to demonstrate the presence of these cells within the tumor microenvironment, which is associated with their tropism for sites of inflammation; however, their role here needs to be clarified. In different investigations, the feasibility of using MSCs or their secretome to treat cancer has been sought, with these results being ambiguous. It has been described that MSCs can be activated and present various phenotypes, which could explain the divergence in their action; however, these activation mechanisms and the different phenotypes still need to be well known. This review explores MSCs and their use in regenerative medicine with a targeted approach to cancer. Impact Statement This text addresses the diverging findings on the role of mesenchymal stem cells in the tumor microenvironment and discrepancies on the use of these cells as cancer treatment, separating the direct use of the cells from the use of the secretome. Multiple authors refer equally to the cells and their secretome to conclude on the positive or negative outcome, without taking into consideration how the cells are affected by their surroundings.
Collapse
Affiliation(s)
- Jhon Alexander Ramírez Idarraga
- Corporación Académica Ciencias Básicas Biomédicas, Universidad de Antioquía, Medellín, Colombia
- Grupo Ingeniería de Tejidos y Terapias Celulares, Instituto de Investigaciones Médicas, Universidad de Antioquía, Medellín, Colombia
| | - Luz Marina Restrepo Múnera
- Grupo Ingeniería de Tejidos y Terapias Celulares, Instituto de Investigaciones Médicas, Universidad de Antioquía, Medellín, Colombia
| |
Collapse
|
13
|
Rivera-Cruz CM, Kumar S, Figueiredo ML. Poly I:C-priming of adipose-derived mesenchymal stromal cells promotes a pro-tumorigenic phenotype in an immunocompetent mouse model of prostate cancer. Front Cell Dev Biol 2023; 11:1145421. [PMID: 38078010 PMCID: PMC10703370 DOI: 10.3389/fcell.2023.1145421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/02/2023] [Indexed: 02/12/2024] Open
Abstract
Introduction: Mesenchymal stromal cells (MSC) are envisioned as a potential cellular vehicle for targeted cancer therapies due to their tumor tropism and immune permissiveness. An obstacle in their use is the duality in their interactions within tumors, rendering them pro-tumorigenic or anti-tumorigenic, in a context dependent manner. MSC preconditioning, or priming, has been proposed as a strategy for directing the effector properties of MSC at tumor sites. Methods: We primed human MSC derived from adipose tissues (ASC), a clinically advantageous MSC source, utilizing toll-like receptor agonists. Subsequently, we explored the consequences in tumor progression and transcriptome upon the interaction of tumor cells with primed or unprimed ASC in an in vivo model of prostate cancer, the second most common cancer and second leading cause of cancer related death in men in the USA. Results and discussion: In the studied model, poly I:C-primed ASC were found to significantly accelerate tumor growth progression. And while unprimed and LPS-primed ASC did not exert a significant effect on tumor growth at the macroscopic level, gene expression analyses suggested that all treatments promoted distinct modulatory effects in the tumor microenvironment, including altered modulation of angiogenesis, and immune response processes. However, the effects resulting from the collective interaction across these processes must be sufficiently skewed in a pro-tumorigenic or anti-tumorigenic direction for evidence of tumor progression modulation to be detectable at the macroscopic level. Our study highlights potential MSC-tumor microenvironment interactions that may be leveraged and should be considered in the development of cancer therapeutics utilizing MSC.
Collapse
Affiliation(s)
| | | | - Marxa L. Figueiredo
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
14
|
Afkhami H, Mahmoudvand G, Fakouri A, Shadab A, Mahjoor M, Komeili Movahhed T. New insights in application of mesenchymal stem cells therapy in tumor microenvironment: pros and cons. Front Cell Dev Biol 2023; 11:1255697. [PMID: 37849741 PMCID: PMC10577325 DOI: 10.3389/fcell.2023.1255697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 09/11/2023] [Indexed: 10/19/2023] Open
Abstract
Multipotent mesenchymal stem cells (MSCs) are widely accepted as a useful tool for cell-based therapy of various diseases including malignancies. The therapeutic effects of MSCs are mainly attributed to their immunomodulatory and immunosuppressive properties. Despite the promising outcomes of MSCs in cancer therapy, a growing body of evidence implies that MSCs also show tumorigenic properties in the tumor microenvironment (TME), which might lead to tumor induction and progression. Owing to the broad-spectrum applications of MSCs, this challenge needs to be tackled so that they can be safely utilized in clinical practice. Herein, we review the diverse activities of MSCs in TME and highlight the potential methods to convert their protumorigenic characteristics into onco-suppressive effects.
Collapse
Affiliation(s)
- Hamed Afkhami
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Golnaz Mahmoudvand
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Arshia Fakouri
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Alireza Shadab
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Iran University of Medical Sciences, Deputy of Health, Tehran, Iran
| | - Mohamad Mahjoor
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
15
|
Slama Y, Ah-Pine F, Khettab M, Arcambal A, Begue M, Dutheil F, Gasque P. The Dual Role of Mesenchymal Stem Cells in Cancer Pathophysiology: Pro-Tumorigenic Effects versus Therapeutic Potential. Int J Mol Sci 2023; 24:13511. [PMID: 37686315 PMCID: PMC10488262 DOI: 10.3390/ijms241713511] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are multipotent cells involved in numerous physiological events, including organogenesis, the maintenance of tissue homeostasis, regeneration, or tissue repair. MSCs are increasingly recognized as playing a major, dual, and complex role in cancer pathophysiology through their ability to limit or promote tumor progression. Indeed, these cells are known to interact with the tumor microenvironment, modulate the behavior of tumor cells, influence their functions, and promote distant metastasis formation through the secretion of mediators, the regulation of cell-cell interactions, and the modulation of the immune response. This dynamic network can lead to the establishment of immunoprivileged tissue niches or the formation of new tumors through the proliferation/differentiation of MSCs into cancer-associated fibroblasts as well as cancer stem cells. However, MSCs exhibit also therapeutic effects including anti-tumor, anti-proliferative, anti-inflammatory, or anti-oxidative effects. The therapeutic interest in MSCs is currently growing, mainly due to their ability to selectively migrate and penetrate tumor sites, which would make them relevant as vectors for advanced therapies. Therefore, this review aims to provide an overview of the double-edged sword implications of MSCs in tumor processes. The therapeutic potential of MSCs will be reviewed in melanoma and lung cancers.
Collapse
Affiliation(s)
- Youssef Slama
- Unité de Recherche Études Pharmaco-Immunologiques (EPI), Université de La Réunion, CHU de La Réunion, Allée des Topazes, 97400 Saint-Denis, La Réunion, France; (F.A.-P.); (M.K.); (P.G.)
- Service de Radiothérapie, Clinique Sainte-Clotilde, Groupe Clinifutur, 127 Route de Bois de Nèfles, 97400 Saint-Denis, La Réunion, France; (M.B.); (F.D.)
- Laboratoire Interdisciplinaire de Recherche en Santé (LIRS), RunResearch, Clinique Sainte-Clotilde, 127 Route de Bois de Nèfles, 97400 Saint-Denis, La Réunion, France;
| | - Franck Ah-Pine
- Unité de Recherche Études Pharmaco-Immunologiques (EPI), Université de La Réunion, CHU de La Réunion, Allée des Topazes, 97400 Saint-Denis, La Réunion, France; (F.A.-P.); (M.K.); (P.G.)
- Service d’Anatomie et Cytologie Pathologiques, CHU de La Réunion sites SUD—Saint-Pierre, Avenue François Mitterrand, 97448 Saint-Pierre Cedex, La Réunion, France
| | - Mohamed Khettab
- Unité de Recherche Études Pharmaco-Immunologiques (EPI), Université de La Réunion, CHU de La Réunion, Allée des Topazes, 97400 Saint-Denis, La Réunion, France; (F.A.-P.); (M.K.); (P.G.)
- Service d’Oncologie Médicale, CHU de La Réunion sites SUD—Saint-Pierre, Avenue François Mitterrand, 97448 Saint-Pierre Cedex, La Réunion, France
| | - Angelique Arcambal
- Laboratoire Interdisciplinaire de Recherche en Santé (LIRS), RunResearch, Clinique Sainte-Clotilde, 127 Route de Bois de Nèfles, 97400 Saint-Denis, La Réunion, France;
| | - Mickael Begue
- Service de Radiothérapie, Clinique Sainte-Clotilde, Groupe Clinifutur, 127 Route de Bois de Nèfles, 97400 Saint-Denis, La Réunion, France; (M.B.); (F.D.)
- Laboratoire Interdisciplinaire de Recherche en Santé (LIRS), RunResearch, Clinique Sainte-Clotilde, 127 Route de Bois de Nèfles, 97400 Saint-Denis, La Réunion, France;
| | - Fabien Dutheil
- Service de Radiothérapie, Clinique Sainte-Clotilde, Groupe Clinifutur, 127 Route de Bois de Nèfles, 97400 Saint-Denis, La Réunion, France; (M.B.); (F.D.)
- Laboratoire Interdisciplinaire de Recherche en Santé (LIRS), RunResearch, Clinique Sainte-Clotilde, 127 Route de Bois de Nèfles, 97400 Saint-Denis, La Réunion, France;
| | - Philippe Gasque
- Unité de Recherche Études Pharmaco-Immunologiques (EPI), Université de La Réunion, CHU de La Réunion, Allée des Topazes, 97400 Saint-Denis, La Réunion, France; (F.A.-P.); (M.K.); (P.G.)
| |
Collapse
|
16
|
Renaud M, Bousquet P, Macias G, Rochefort GY, Durand JO, Marsal LF, Cuisinier F, Cunin F, Collart-Dutilleul PY. Allogenic Stem Cells Carried by Porous Silicon Scaffolds for Active Bone Regeneration In Vivo. Bioengineering (Basel) 2023; 10:852. [PMID: 37508879 PMCID: PMC10376284 DOI: 10.3390/bioengineering10070852] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
To date, bone regeneration techniques use many biomaterials for bone grafting with limited efficiencies. For this purpose, tissue engineering combining biomaterials and stem cells is an important avenue of development to improve bone regeneration. Among potentially usable non-toxic and bioresorbable scaffolds, porous silicon (pSi) is an interesting biomaterial for bone engineering. The possibility of modifying its surface can allow a better cellular adhesion as well as a control of its rate of resorption. Moreover, release of silicic acid upon resorption of its nanostructure has been previously proved to enhance stem cell osteodifferentiation by inducing calcium phosphate formation. In the present study, we used a rat tail model to experiment bone tissue engineering with a critical size defect. Two groups with five rats per group of male Wistar rats were used. In each rat, four vertebrae were used for biomaterial implantation. Randomized bone defects were filled with pSi particles alone or pSi particles carrying dental pulp stem cells (DPSC). Regeneration was evaluated in comparison to empty defect and defects filled with xenogenic bone substitute (Bio-Oss®). Fluorescence microscopy and SEM evaluations showed adhesion of DPSCs on pSi particles with cells exhibiting distribution throughout the biomaterial. Histological analyzes revealed the formation of a collagen network when the defects were filled with pSi, unlike the positive control using Bio-Oss®. Overall bone formation was objectivated with µCT analysis and showed a higher bone mineral density with pSi particles combining DPSC. Immunohistochemical assays confirmed the increased expression of bone markers (osteocalcin) when pSi particles carried DPSC. Surprisingly, no grafted cells remained in the regenerated area after one month of healing, even though the grafting of DPSC clearly increased bone regeneration for both bone marker expression and overall bone formation objectivated with µCT. In conclusion, our results show that the association of pSi with DPSCs in vivo leads to greater bone formation, compared to a pSi graft without DPSCs. Our results highlight the paracrine role of grafted stem cells by recruitment and stimulation of endogenous cells.
Collapse
Affiliation(s)
- Matthieu Renaud
- Laboratoire Biosanté et Nanoscience (LBN), Université Montpellier, 34000 Montpellier, France; (M.R.); (F.C.)
- Faculty of Dentistry, Université de Tours, 37000 Tours, France
| | - Philippe Bousquet
- Laboratoire Biosanté et Nanoscience (LBN), Université Montpellier, 34000 Montpellier, France; (M.R.); (F.C.)
- Faculty of Dentistry, Université Montpellier, 34000 Montpellier, France
- Service Odontologie, Hospital Center University de Montpellier, 34000 Montpellier, France
| | - Gerard Macias
- Institute Charles Gerhardt Montpellier (ICGM), Université Montpellier, Centre National de la Recherche Scientifique (CNRS), ENSCM, 34000 Montpellier, France
- Department of Electronic, Electrical and Automatic Engineering (DEEEA), Universitat Rovira i Virgili, 43003 Tarragona, Spain
| | | | - Jean-Olivier Durand
- Institute Charles Gerhardt Montpellier (ICGM), Université Montpellier, Centre National de la Recherche Scientifique (CNRS), ENSCM, 34000 Montpellier, France
| | - Lluis F. Marsal
- Department of Electronic, Electrical and Automatic Engineering (DEEEA), Universitat Rovira i Virgili, 43003 Tarragona, Spain
| | - Frédéric Cuisinier
- Laboratoire Biosanté et Nanoscience (LBN), Université Montpellier, 34000 Montpellier, France; (M.R.); (F.C.)
- Faculty of Dentistry, Université Montpellier, 34000 Montpellier, France
- Service Odontologie, Hospital Center University de Montpellier, 34000 Montpellier, France
| | - Frédérique Cunin
- Institute Charles Gerhardt Montpellier (ICGM), Université Montpellier, Centre National de la Recherche Scientifique (CNRS), ENSCM, 34000 Montpellier, France
| | - Pierre-Yves Collart-Dutilleul
- Laboratoire Biosanté et Nanoscience (LBN), Université Montpellier, 34000 Montpellier, France; (M.R.); (F.C.)
- Faculty of Dentistry, Université Montpellier, 34000 Montpellier, France
- Service Odontologie, Hospital Center University de Montpellier, 34000 Montpellier, France
| |
Collapse
|
17
|
Furusaka Y, Inoue S, Mizoguchi I, Hasegawa H, Katahira Y, Watanabe A, Sakamoto E, Sekine A, Miyakawa S, Umezu T, Owaki T, Yoneto T, Yoshimoto T. Potent antitumor effects of the conditioned medium of bone marrow-derived mesenchymal stem cells via IGFBP-4. Cancer Sci 2023; 114:2499-2514. [PMID: 36942841 PMCID: PMC10486206 DOI: 10.1111/cas.15789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 03/23/2023] Open
Abstract
Cell transfer therapy using mesenchymal stem cells (MSCs) has pronounced therapeutic potential, but concerns remain about immune rejection, emboli formation, and promotion of tumor progression. Because the mode of action of MSCs highly relies on their paracrine effects through secretion of bioactive molecules, cell-free therapy using the conditioned medium (CM) of MSCs is an attractive option. However, the effects of MSC-CM on tumor progression have not been fully elucidated. Herein, we addressed this issue and investigated the possible underlying molecular mechanisms. The CM of MSCs derived from human bone marrow greatly inhibited the in vitro growth of several human tumor cell lines and the in vivo growth of the SCCVII murine squamous cell carcinoma cell line with reduced neovascularization. Exosomes in the MSC-CM were only partially involved in the inhibitory effects. The CM contained a variety of cytokines including insulin-like growth factor binding proteins (IGFBPs). Among them, IGFBP-4 greatly inhibited the in vitro growth of these tumors and angiogenesis, and immunodepletion of IGFBP-4 from the CM significantly reversed these effects. Of note, the CM greatly reduced the phosphorylation of AKT, ERK, IGF-1 receptor beta, and p38 MAPK in a partly IGFBP4-dependent manner, possibly through its binding to IGF-1/2 and blocking the signaling. The CM depleted of IGFBP-4 also reversed the inhibitory effects on in vivo tumor growth and neovascularization. Thus, MSC-CM has potent inhibitory effects on tumor growth and neovascularization in an IGFBP4-dependent manner, suggesting that cell-free therapy using MSC-CM could be a safer promising alternative for even cancer patients.
Collapse
Affiliation(s)
- Yuma Furusaka
- Department of Immunoregulation, Institute of Medical ScienceTokyo Medical University6‐1‐1 Shinjuku, Shinjuku‐kuTokyo160‐8402Japan
| | - Shinya Inoue
- Department of Immunoregulation, Institute of Medical ScienceTokyo Medical University6‐1‐1 Shinjuku, Shinjuku‐kuTokyo160‐8402Japan
| | - Izuru Mizoguchi
- Department of Immunoregulation, Institute of Medical ScienceTokyo Medical University6‐1‐1 Shinjuku, Shinjuku‐kuTokyo160‐8402Japan
| | - Hideaki Hasegawa
- Department of Immunoregulation, Institute of Medical ScienceTokyo Medical University6‐1‐1 Shinjuku, Shinjuku‐kuTokyo160‐8402Japan
| | - Yasuhiro Katahira
- Department of Immunoregulation, Institute of Medical ScienceTokyo Medical University6‐1‐1 Shinjuku, Shinjuku‐kuTokyo160‐8402Japan
| | - Aruma Watanabe
- Department of Immunoregulation, Institute of Medical ScienceTokyo Medical University6‐1‐1 Shinjuku, Shinjuku‐kuTokyo160‐8402Japan
| | - Eri Sakamoto
- Department of Immunoregulation, Institute of Medical ScienceTokyo Medical University6‐1‐1 Shinjuku, Shinjuku‐kuTokyo160‐8402Japan
| | - Ami Sekine
- Department of Immunoregulation, Institute of Medical ScienceTokyo Medical University6‐1‐1 Shinjuku, Shinjuku‐kuTokyo160‐8402Japan
| | - Satomi Miyakawa
- Department of Immunoregulation, Institute of Medical ScienceTokyo Medical University6‐1‐1 Shinjuku, Shinjuku‐kuTokyo160‐8402Japan
| | - Tomohiro Umezu
- Department of Molecular PathologyTokyo Medical University6‐1‐1 Shinjuku, Shinjuku‐kuTokyo160‐8402Japan
| | - Toshiyuki Owaki
- Department of Immunoregulation, Institute of Medical ScienceTokyo Medical University6‐1‐1 Shinjuku, Shinjuku‐kuTokyo160‐8402Japan
| | - Toshihiko Yoneto
- Department of Immunoregulation, Institute of Medical ScienceTokyo Medical University6‐1‐1 Shinjuku, Shinjuku‐kuTokyo160‐8402Japan
| | - Takayuki Yoshimoto
- Department of Immunoregulation, Institute of Medical ScienceTokyo Medical University6‐1‐1 Shinjuku, Shinjuku‐kuTokyo160‐8402Japan
| |
Collapse
|
18
|
Aru B, Akdeniz T, Dağdeviren H, Gürel G, Yanıkkaya Demirel G. Testosterone Propionate Promotes Proliferation and Viability of Bone Marrow Mesenchymal Stem Cells while Preserving Their Characteristics and Inducing Their Anti-Cancer Efficacy. Balkan Med J 2023; 40:117-123. [PMID: 36748249 PMCID: PMC9998833 DOI: 10.4274/balkanmedj.galenos.2022.2022-10-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Background Various studies have reported the effects of testosterone on different cell types, yet bone marrow-derived mesenchymal stem cells’ cellular responses to testosterone remain unknown. Aims To investigate the effects of testosterone propionate, an oil-soluble short-acting form of testosterone, on human bone marrow-derived mesenchymal stem cells’ proliferation and viability after 24 hours of incubation. We also investigated the impact of testosterone propionate on bone marrow-derived mesenchymal stem cell’s polarization and cytotoxicity on K562 leukemia cell line. Study Design In vitro study. Methods We expanded commercially available bone marrow derived mesenchymal stem cells in vitro and treated them with testosterone propionate at concentrations ranging from 10-6-10-10 M for 24 hours. Ideal concentration was determined by evaluating cellular viability and proliferation with Annexin V/Propidium Iodide assay and carboxyfluorescein succinimidyl ester staining. The characteristic features of bone marrow-derived mesenchymal stem cells were evaluated by immunophenotyping and investigating their differentiation capacities. Bone marrow-derived mesenchymal stem cells’ cytotoxic properties upon testosterone propionate treatment were determined by co-culturing the cells with K562 cells and with confocal imaging investigating polarization. Results Testosterone propionate promoted proliferation and maintained the viability of bone marrow-derived mesenchymal stem at 10-8 M concentration. Further evaluations were conducted with the determined dose. The results showed that, apart from promoting mesenchymal stem cells’ polarization and increasing their cytotoxicity on K562 cells, testosterone propionate did not alter differentiation capacities of bone marrow-derived mesenchymal stem cells and certain cell surface markers, but led to a significant increase in HLA-DR expression. Conclusion The findings reveal that testosterone propionate promotes the proliferation and survival of bone marrow-derived mesenchymal stem cells in a dose-dependent manner without hampering their differentiation capacities, induces their polarization to the pro-inflammatory phenotype, and increases their cytotoxicity on the K562 cell line.
Collapse
Affiliation(s)
- Başak Aru
- Department of Immunology, Yeditepe University Faculty of Medicine, İstanbul, Turkey
| | - Tuba Akdeniz
- Department of Medical Biology, Yeditepe University Faculty of Medicine, İstanbul, Turkey
| | - Hüsniye Dağdeviren
- Stem Cell Laboratory, Yeditepe University Training and Research Hospital, İstanbul, Turkey
| | - Gizem Gürel
- Department of Immunology, Yeditepe University Faculty of Medicine, İstanbul, Turkey
| | - Gülderen Yanıkkaya Demirel
- Department of Immunology, Yeditepe University Faculty of Medicine, İstanbul, Turkey,Stem Cell Laboratory, Yeditepe University Training and Research Hospital, İstanbul, Turkey
| |
Collapse
|
19
|
Samadi A, Moammeri A, Pourmadadi M, Abbasi P, Hosseinpour Z, Farokh A, Shamsabadipour A, Heydari M, Mohammadi MR. Cell Encapsulation and 3D Bioprinting for Therapeutic Cell Transplantation. ACS Biomater Sci Eng 2023; 9:1862-1890. [PMID: 36877212 DOI: 10.1021/acsbiomaterials.2c01183] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
The promise of cell therapy has been augmented by introducing biomaterials, where intricate scaffold shapes are fabricated to accommodate the cells within. In this review, we first discuss cell encapsulation and the promising potential of biomaterials to overcome challenges associated with cell therapy, particularly cellular function and longevity. More specifically, cell therapies in the context of autoimmune disorders, neurodegenerative diseases, and cancer are reviewed from the perspectives of preclinical findings as well as available clinical data. Next, techniques to fabricate cell-biomaterials constructs, focusing on emerging 3D bioprinting technologies, will be reviewed. 3D bioprinting is an advancing field that enables fabricating complex, interconnected, and consistent cell-based constructs capable of scaling up highly reproducible cell-biomaterials platforms with high precision. It is expected that 3D bioprinting devices will expand and become more precise, scalable, and appropriate for clinical manufacturing. Rather than one printer fits all, seeing more application-specific printer types, such as a bioprinter for bone tissue fabrication, which would be different from a bioprinter for skin tissue fabrication, is anticipated in the future.
Collapse
Affiliation(s)
- Amirmasoud Samadi
- Department of Chemical and Biomolecular Engineering, 6000 Interdisciplinary Science & Engineering Building (ISEB), Irvine, California 92617, United States
| | - Ali Moammeri
- School of Chemical Engineering, College of Engineering, University of Tehran, Enghelab Square, 16 Azar Street, Tehran 1417935840, Iran
| | - Mehrab Pourmadadi
- School of Chemical Engineering, College of Engineering, University of Tehran, Enghelab Square, 16 Azar Street, Tehran 1417935840, Iran
| | - Parisa Abbasi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Azadi Avenue, Tehran 1458889694, Iran
| | - Zeinab Hosseinpour
- Biotechnology Research Laboratory, Faculty of Chemical Engineering, Babol Noshirvani University of Technology, Babol 4714871167, Mazandaran Province, Iran
| | - Arian Farokh
- School of Chemical Engineering, College of Engineering, University of Tehran, Enghelab Square, 16 Azar Street, Tehran 1417935840, Iran
| | - Amin Shamsabadipour
- School of Chemical Engineering, College of Engineering, University of Tehran, Enghelab Square, 16 Azar Street, Tehran 1417935840, Iran
| | - Maryam Heydari
- Department of Cell and Molecular Biology, Faculty of Biological Science, University of Kharazmi, Tehran 199389373, Iran
| | - M Rezaa Mohammadi
- Dale E. and Sarah Ann Fowler School of Engineering, Chapman University, Orange, California 92866, United States
| |
Collapse
|
20
|
Oncologic Safety and Efficacy of Cell-Assisted Lipotransfer for Breast Reconstruction in a Murine Model of Residual Breast Cancer. Aesthetic Plast Surg 2023; 47:412-422. [PMID: 35918436 DOI: 10.1007/s00266-022-03021-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/08/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Cell-assisted lipotransfer (CAL) is a novel technique for fat grafting that combines the grafting of autologous fat and adipose-derived stromal cells (ASCs) to enhance fat graft retention; however, its oncologic safety is controversial. METHODS Herein, we investigated the oncologic safety of CAL for breast reconstruction using a murine model of residual breast cancer. Various concentrations of 4T1 cells (murine breast cancer cells) were injected into female mastectomized BALB/c mice to determine the appropriate concentration for injection. One week after injection, mice were divided into control (100 μL fat), low CAL (2.5 × 105 ASCs/100 μL fat), and high CAL (1.0 × 106 ASCs/100 μL fat) groups, and fat grafting was performed. The injection of 5.0 × 103 4T1 cells was appropriate to produce a murine model of residual breast cancer. RESULTS The weight of the fat tumor mass was significantly higher in the high CAL group than in the other groups (p < 0.05). However, the estimated tumor weight was not significantly different between the groups. Additionally, the fat graft survival rate was significantly higher in the high CAL group than in the control and low CAL groups (p < 0.05). No significant difference was noted in the percentage of Ki-67-positive cells, suggesting that tumor proliferation was not significantly different between the groups. CONCLUSION In summary, CAL significantly improved fat graft survival without affecting tumor size and proliferation in a murine model of residual breast cancer. These results highlight the oncologic safety of CAL for breast reconstruction. NO LEVEL ASSIGNED This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
|
21
|
Rivera-Cruz CM, Figueiredo ML. Evaluation of human adipose-derived mesenchymal stromal cell Toll-like receptor priming and effects on interaction with prostate cancer cells. Cytotherapy 2023; 25:33-45. [PMID: 36257875 DOI: 10.1016/j.jcyt.2022.09.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 07/20/2022] [Accepted: 09/26/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND AIMS Mesenchymal stromal cells (MSCs) are a multipotent cell population of clinical interest because of their ability to migrate to injury and tumor sites, where they may participate in tissue repair and modulation of immune response. Although the processes regulating MSC function are incompletely understood, it has been shown that stimulation of Toll-like receptors (TLRs) can alter MSC activity. More specifically, it has been reported that human bone marrow-derived MSCs can be "polarized" by TLR priming into contrasting immunomodulatory functions, with opposite (supportive or suppressive) roles in tumor progression and inflammation. Adipose-derived MSCs (ASCs) represent a promising alternative MSC subpopulation for therapeutic development because of their relative ease of isolation and higher abundance compared with their bone marrow-derived counterparts; however, the polarization of ASCs remains unreported. METHODS In this study, we evaluated the phenotypic and functional consequences of short-term, low-level stimulation of ASCs with TLR3 and TLR4 agonists. RESULTS In these assays, we identified transient gene expression changes resembling the reported pro-inflammatory and anti-inflammatory MSC phenotypes. Furthermore, these priming strategies led to changes in the functional properties of ASCs, affecting their ability to migrate and modulate immune-mediated responses to prostate cancer cells in vitro. CONCLUSIONS TLR3 stimulation significantly decreased ASC migration, and TLR4 stimulation increased ASC immune-mediated killing potential against prostate cancer cells.
Collapse
Affiliation(s)
- Cosette M Rivera-Cruz
- Department of Basic Medical Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Marxa L Figueiredo
- Department of Basic Medical Sciences, Purdue University, West Lafayette, Indiana, USA.
| |
Collapse
|
22
|
Wang Z, Sun Y, Shen R, Tang X, Xu Y, Zhang Y, Liu Y. Global scientific trends on the immunomodulation of mesenchymal stem cells in the 21st century: A bibliometric and visualized analysis. Front Immunol 2022; 13:984984. [PMID: 36090982 PMCID: PMC9449834 DOI: 10.3389/fimmu.2022.984984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 08/05/2022] [Indexed: 11/15/2022] Open
Abstract
Background Since the discovery of the immunomodulatory functions of mesenchymal stem cells (MSCs), their application in immunomodulation has attracted considerable attention, and an increasing number of studies have been conducted worldwide. Our research aimed to investigate the global status and trends in this field. Methods Publications on the immunomodulatory functions of MSCs from 1 January 2000 to 7 March 2022 were retrieved from the Web of Science Core Collection. The data were studied and indexed using the bibliometric methodology. Visualization analysis, co-authorship, co-occurrence analysis, and publication trends in MSC immunomodulation were conducted using the VOSviewer software. Results In total, 4,227 papers were included in the study. The number of publications and research interests has significantly increased globally. China published the highest number of related articles, while the US published articles with the highest number of citations. Stem Cell Research & Therapy had the highest number of publications. Sun Yat-sen University, Shanghai Jiao Tong University, Harvard University, and Seoul National University were the most contributive institutions. Furthermore, the studies were divided into four research hotspots for MSC immunomodulation: MSC immunomodulation in regenerative medicine, the effects and mechanisms of MSC immunomodulation, MSC therapy for immune diseases, and the cell source of MSCs. Conclusion This study indicates that the number of publications on MSC immunomodulation will increase in the future, and MSC immunomodulation mechanisms and clinical applications of MSC immunotherapy should be the next hotspots in this research field.
Collapse
Affiliation(s)
- Zhongqing Wang
- Department of Information Center, The First Hospital of China Medical University, Shenyang, China
| | - Yuqiang Sun
- Department of Emergency, The First Hospital of China Medical University, Shenyang, China
| | - Rou Shen
- Department of Pediatric Dentistry, School and Hospital of Stomatology, China Medical University, Shenyang, China
- Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Xia Tang
- Department of Pediatric Dentistry, School and Hospital of Stomatology, China Medical University, Shenyang, China
- Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Yingxin Xu
- Department of Information Center, The First Hospital of China Medical University, Shenyang, China
| | - Ye Zhang
- Department of Information Center, The First Hospital of China Medical University, Shenyang, China
| | - Yao Liu
- Department of Pediatric Dentistry, School and Hospital of Stomatology, China Medical University, Shenyang, China
- Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
- *Correspondence: Yao Liu,
| |
Collapse
|
23
|
Han HT, Jin WL, Li X. Mesenchymal stem cells-based therapy in liver diseases. MOLECULAR BIOMEDICINE 2022; 3:23. [PMID: 35895169 PMCID: PMC9326420 DOI: 10.1186/s43556-022-00088-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/20/2022] [Indexed: 12/24/2022] Open
Abstract
Multiple immune cells and their products in the liver together form a complex and unique immune microenvironment, and preclinical models have demonstrated the importance of imbalances in the hepatic immune microenvironment in liver inflammatory diseases and immunocompromised liver diseases. Various immunotherapies have been attempted to modulate the hepatic immune microenvironment for the purpose of treating liver diseases. Mesenchymal stem cells (MSCs) have a comprehensive and plastic immunomodulatory capacity. On the one hand, they have been tried for the treatment of inflammatory liver diseases because of their excellent immunosuppressive capacity; On the other hand, MSCs have immune-enhancing properties in immunocompromised settings and can be modified into cellular carriers for targeted transport of immune enhancers by genetic modification, physical and chemical loading, and thus they are also used in the treatment of immunocompromised liver diseases such as chronic viral infections and hepatocellular carcinoma. In this review, we discuss the immunological basis and recent strategies of MSCs for the treatment of the aforementioned liver diseases. Specifically, we update the immune microenvironment of the liver and summarize the distinct mechanisms of immune microenvironment imbalance in inflammatory diseases and immunocompromised liver diseases, and how MSCs can fully exploit their immunotherapeutic role in liver diseases with both immune imbalance patterns.
Collapse
Affiliation(s)
- Heng-Tong Han
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, P. R, China
| | - Wei-Lin Jin
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, P. R, China
- Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, No. 1 West Donggang Road, Lanzhou, 730000, People's Republic of China
| | - Xun Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, P. R, China.
- Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, No. 1 West Donggang Road, Lanzhou, 730000, People's Republic of China.
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China.
- Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, 730000, People's Republic of China.
| |
Collapse
|
24
|
Mesenchymal stem cells: A living carrier for active tumor-targeted delivery. Adv Drug Deliv Rev 2022; 185:114300. [PMID: 35447165 DOI: 10.1016/j.addr.2022.114300] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 03/22/2022] [Accepted: 04/12/2022] [Indexed: 12/16/2022]
Abstract
The strategy of using mesenchymal stem cells (MSCs) as a living carrier for active delivery of therapeutic agents targeting tumor sites has been attempted in a wide range of studies to validate the feasibility and efficacy for tumor treatment. This approach reveals powerful tumor targeting and tumor penetration. In addition, MSCs have been confirmed to actively participate in immunomodulation of the tumor microenvironment. Thus, MSCs are not inert delivery vehicles but have a strong impact on the fate of tumor cells. In this review, these active properties of MSCs are addressed to highlight the advantages and challenges of using MSCs for tumor-targeted delivery. In addition, some of the latest examples of using MSCs to carry a variety of anti-tumor agents for tumor-targeted therapy are summarized. Recent technologies to improve the performance and safety of this delivery strategy will be introduced. The advances, applications, and challenges summarized in this review will provide a general understanding of this promising strategy for actively delivering drugs to tumor tissues.
Collapse
|
25
|
Zhang L, Ma XJN, Fei YY, Han HT, Xu J, Cheng L, Li X. Stem cell therapy in liver regeneration: Focus on mesenchymal stem cells and induced pluripotent stem cells. Pharmacol Ther 2022; 232:108004. [PMID: 34597754 DOI: 10.1016/j.pharmthera.2021.108004] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/11/2021] [Accepted: 09/23/2021] [Indexed: 02/07/2023]
Abstract
The liver has the ability to repair itself after injury; however, a variety of pathological changes in the liver can affect its ability to regenerate, and this could lead to liver failure. Mesenchymal stem cells (MSCs) are considered a good source of cells for regenerative medicine, as they regulate liver regeneration through different mechanisms, and their efficacy has been demonstrated by many animal experiments and clinical studies. Induced pluripotent stem cells, another good source of MSCs, have also made great progress in the establishment of organoids, such as liver disease models, and in drug screening. Owing to the recent developments in MSCs and induced pluripotent stem cells, combined with emerging technologies including graphene, nano-biomaterials, and gene editing, precision medicine and individualized clinical treatment may be realized in the near future.
Collapse
Affiliation(s)
- Lu Zhang
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, PR China; Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, PR China; The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China
| | - Xiao-Jing-Nan Ma
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China
| | - Yuan-Yuan Fei
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, PR China; Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, PR China
| | - Heng-Tong Han
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China
| | - Jun Xu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China
| | - Lu Cheng
- Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, PR China
| | - Xun Li
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, PR China; Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, PR China; Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, Lanzhou 730000, PR China; Hepatopancreatobiliary Surgery Institute of Gansu Province, Lanzhou 730000, PR China; The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China.
| |
Collapse
|
26
|
[Research progress of external volume expansion assisted autologous fat grafting for breast reconstruction]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2022; 36:370-375. [PMID: 35293180 PMCID: PMC8923929 DOI: 10.7507/1002-1892.202111016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
OBJECTIVE To review the application progress, mechanism, application points, limitations, and oncological safety of external volume expansion (EVE) assisted autologous fat grafting for breast reconstruction and provide a reference for optimizing the design of EVE. METHODS Based on the latest relevant articles, the basic experiments and clinical applications of EVE were summarized. RESULTS EVE can reduce interstitial fluid pressure, increase blood supply, and promote adipogenic differentiation, thereby benefiting the survival of transplanted fat. EVE assisted autologous fat grafting in clinical practice can improve the retention rate of breast volume and the outcome of breast reconstruction, meanwhile it doesn't increase the risk of local recurrence. But there is no standard parameters for application, and there are many complications and limitations. CONCLUSION EVE improves the survival of transplanted fat, but its complications and poor compliance are obvious, so it is urgent to further investigate customized products for breast reconstruction after breast cancer and establish relevant application guidelines.
Collapse
|
27
|
Nii T, Tabata Y. Immunosuppressive mesenchymal stem cells aggregates incorporating hydrogel microspheres promote an in vitro invasion of cancer cells. Regen Ther 2022; 18:516-522. [PMID: 34977285 PMCID: PMC8668441 DOI: 10.1016/j.reth.2021.11.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/16/2021] [Accepted: 11/24/2021] [Indexed: 12/20/2022] Open
Abstract
Introduction The objective of this study is to design a co-culture system of cancer cells and three-dimensional (3D) mesenchymal stem cells (MSC) aggregates for the in vitro evaluation of cancer invasion. Methods First, the MSC of an immunosuppressive phenotype (MSC2) were prepared by the MSC stimulation of polyriboinosinic polyribocytidylic acid. By simple mixing MSC2 and gelatin hydrogel microspheres (GM) in a U-bottomed well of 96 well plates which had been pre-coated with poly (vinyl alcohol), 3D MSC2 aggregates incorporating GM were obtained. The amount of chemokine (C–C motif) ligand 5 (CCL5) secreted from the MSC2 aggregates incorporating GM. Finally, an invasion assay was performed to evaluate the cancer invasion rate by co-cultured cancer cells and the 3D MSC2 incorporating GM. Results The amount of CCL5 secreted for the 3D MSC2 aggregates incorporating GM was significantly higher than that of two-dimensional (2D) MSC, 2D MSC2, and 3D MSC aggregates incorporating GM. When MDA-MB-231 human breast cancer cells were co-cultured with the 3D MSC2 aggregates incorporating GM, the invasion rate of cancer cells was significantly high compared with that of 2D MSC or 2D MSC2 and 3D MSC aggregates incorporating GM. In addition, high secretion of matrix metalloproteinase-2 was observed for the 3D MSC2 aggregates/cancer cells system. Conclusions It is concluded that the co-culture system of 3D MSC2 aggregates incorporating GM and cancer cells is promising to evaluate the invasion of cancer cells in vitro. This invasion model is an important tool for anti-cancer drug screening. Mesenchymal stem cells of an immunosuppressive phenotype (MSC2) were obtained. 3D MSC2 aggregates incorporating gelatin hydrogel microspheres were prepared. 3D MSC2 aggregates promoted the invasion rate of cancer cells.
Collapse
Key Words
- (CCL)5, chemokine (C–C motif) ligand
- 2D, two-dimensional
- 3D, three-dimensional
- Anti-cancer drug screening
- CAF, cancer-associated fibroblasts
- Cancer invasion model
- DDW, double-distilled water
- DMEM, Dulbecco's modified Eagle's medium
- ELISA, enzyme-linked immunosolvent assay
- FCS, fetal calf serum
- GM, gelatin hydrogel microspheres
- Gelatin hydrogel microspheres
- MEM, minimum essential medium
- MMP, matrix metalloproteinase
- MSC, mesenchymal stem cells
- MSC2, MSC of an immunosuppressive phenotype
- Mesenchymal stem cells
- PBS, phosphate buffered-saline
- PVA, poly (vinyl alcohol)
- TAM, tumor-associated macrophages
- Three-dimensional cell culture
Collapse
Affiliation(s)
- Teruki Nii
- Laboratory of Biomaterials, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawara-cho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yasuhiko Tabata
- Laboratory of Biomaterials, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawara-cho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
28
|
Xu C, Wang M, Zandieh-Doulabi B, Sun W, Wei L, Liu Y. To B (Bone Morphogenic Protein-2) or Not to B (Bone Morphogenic Protein-2): Mesenchymal Stem Cells May Explain the Protein's Role in Osteosarcomagenesis. Front Cell Dev Biol 2021; 9:740783. [PMID: 34869325 PMCID: PMC8635864 DOI: 10.3389/fcell.2021.740783] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/11/2021] [Indexed: 12/14/2022] Open
Abstract
Osteosarcoma (OS), a primary malignant bone tumor, stems from bone marrow-derived mesenchymal stem cells (BMSCs) and/or committed osteoblast precursors. Distant metastases, in particular pulmonary and skeletal metastases, are common in patients with OS. Moreover, extensive resection of the primary tumor and bone metastases usually leads to bone defects in these patients. Bone morphogenic protein-2 (BMP-2) has been widely applied in bone regeneration with the rationale that BMP-2 promotes osteoblastic differentiation of BMSCs. Thus, BMP-2 might be useful after OS resection to repair bone defects. However, the potential tumorigenicity of BMP-2 remains a concern that has impeded the administration of BMP-2 in patients with OS and in populations susceptible to OS with severe bone deficiency (e.g., in patients with genetic mutation diseases and aberrant activities of bone metabolism). In fact, some studies have drawn the opposite conclusion about the effect of BMP-2 on OS progression. Given the roles of BMSCs in the origination of OS and osteogenesis, we hypothesized that the responses of BMSCs to BMP-2 in the tumor milieu may be responsible for OS development. This review focuses on the relationship among BMSCs, BMP-2, and OS cells; a better understanding of this relationship may elucidate the accurate mechanisms of actions of BMP-2 in osteosarcomagenesis and thereby pave the way for clinically safer and broader administration of BMP-2 in the future. For example, a low dosage of and a slow-release delivery strategy for BMP-2 are potential topics for exploration to treat OS.
Collapse
Affiliation(s)
- Chunfeng Xu
- Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Mingjie Wang
- Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Behrouz Zandieh-Doulabi
- Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Wei Sun
- Department of Mechanical Engineering, Drexel University, Philadelphia, PA, United States.,Department of Mechanical Engineering, Tsinghua University, Beijing, China
| | - Lingfei Wei
- Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Department of Oral Implantology, Yantai Stomatological Hospital, Yantai, China
| | - Yuelian Liu
- Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
29
|
Xuan X, Tian C, Zhao M, Sun Y, Huang C. Mesenchymal stem cells in cancer progression and anticancer therapeutic resistance. Cancer Cell Int 2021; 21:595. [PMID: 34736460 PMCID: PMC8570012 DOI: 10.1186/s12935-021-02300-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 10/26/2021] [Indexed: 12/26/2022] Open
Abstract
Increasing evidence indicates that the tumor microenvironment appears to play an increasingly important role in cancer progression and therapeutic resistance. Several types of cells within the tumor stroma had distinct impacts on cancer progression, either promoting or inhibiting cancer cell growth. Mesenchymal stem cells (MSCs) are a distinct type of cells that is linked to tumor development. MSCs are recognized for homing to tumor locations and promoting or inhibiting cancer cell proliferation, angiogenesis and metastasis. Moreover, emerging studies suggests that MSCs are also involved in therapeutic resistance. In this review, we analyzed the existing researches and elaborate on the functions of MSCs in cancer progression and anticancer therapeutic resistance, demonstrating that MSCs may be a viable cancer therapeutic target.
Collapse
Affiliation(s)
- Xiuyun Xuan
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Chunxia Tian
- Department of Cardiology, Hubei Provincial Hospital of TCM, Wuhan, 430022, Hubei, China
| | - Mengjie Zhao
- Department of Dermatology, Zhongnan Hospital, Wuhan University, Wuhan, 430022, Hubei, China.
| | - Yanhong Sun
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China.
| | - Changzheng Huang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
| |
Collapse
|
30
|
Tumorigenic Aspects of MSC Senescence-Implication in Cancer Development and Therapy. J Pers Med 2021; 11:jpm11111133. [PMID: 34834485 PMCID: PMC8618265 DOI: 10.3390/jpm11111133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 12/13/2022] Open
Abstract
As an organism ages, many physiological processes change, including the immune system. This process, called immunosenescence, characterized by abnormal activation and imbalance of innate and adaptive immunity, leads to a state of chronic low-grade systemic inflammation, termed inflammaging. Aging and inflammaging are considered to be the root of many diseases of the elderly, as infections, autoimmune and chronic inflammatory diseases, degenerative diseases, and cancer. The role of mesenchymal stromal/stem cells (MSCs) in the inflammaging process and the age-related diseases is not completely established, although numerous features of aging MSCs, including altered immunomodulatory properties, impeded MSC niche supporting functions, and senescent MSC secretory repertoire are consistent with inflammaging development. Although senescence has its physiological function and can represent a mechanism of tumor prevention, in most cases it eventually transforms into a deleterious (para-)inflammatory process that promotes tumor growth. In this review we are going through current literature, trying to explore the role of senescent MSCs in making and/or sustaining a microenvironment permissive to tumor development and to analyze the therapeutic options that could target this process.
Collapse
|
31
|
Manoukian P, Bijlsma M, van Laarhoven H. The Cellular Origins of Cancer-Associated Fibroblasts and Their Opposing Contributions to Pancreatic Cancer Growth. Front Cell Dev Biol 2021; 9:743907. [PMID: 34646829 PMCID: PMC8502878 DOI: 10.3389/fcell.2021.743907] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 08/30/2021] [Indexed: 12/17/2022] Open
Abstract
Pancreatic tumors are known to harbor an abundant and highly desmoplastic stroma. Among the various cell types that reside within tumor stroma, cancer-associated fibroblasts (CAFs) have gained a lot of attention in the cancer field due to their contributions to carcinogenesis and tumor architecture. These cells are not a homogeneous population, but have been shown to have different origins, phenotypes, and contributions. In pancreatic tumors, CAFs generally emerge through the activation and/or recruitment of various cell types, most notably resident fibroblasts, pancreatic stellate cells (PSCs), and tumor-infiltrating mesenchymal stem cells (MSCs). In recent years, single cell transcriptomic studies allowed the identification of distinct CAF populations in pancreatic tumors. Nonetheless, the exact sources and functions of those different CAF phenotypes remain to be fully understood. Considering the importance of stromal cells in pancreatic cancer, many novel approaches have aimed at targeting the stroma but current stroma-targeting therapies have yielded subpar results, which may be attributed to heterogeneity in the fibroblast population. Thus, fully understanding the roles of different subsets of CAFs within the stroma, and the cellular dynamics at play that contribute to heterogeneity in CAF subsets may be essential for the design of novel therapies and improving clinical outcomes. Fortunately, recent advances in technologies such as microfluidics and bio-printing have made it possible to establish more advanced ex vivo models that will likely prove useful. In this review, we will present the different roles of stromal cells in pancreatic cancer, focusing on CAF origin as a source of heterogeneity, and the role this may play in therapy failure. We will discuss preclinical models that could be of benefit to the field and that may contribute to further clinical development.
Collapse
Affiliation(s)
- Paul Manoukian
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Maarten Bijlsma
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Hanneke van Laarhoven
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
32
|
Karimi-Shahri M, Javid H, Sharbaf Mashhad A, Yazdani S, Hashemy SI. Mesenchymal stem cells in cancer therapy; the art of harnessing a foe to a friend. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:1307-1323. [PMID: 35096289 PMCID: PMC8769515 DOI: 10.22038/ijbms.2021.58227.12934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/04/2021] [Indexed: 12/09/2022]
Abstract
For a long time, mesenchymal stem cells (MSCs) were discussed only as stem cells which could give rise to different types of cells. However, when it became clear that their presence in the tumor microenvironment (TME) was like a green light for tumorigenesis, they emerged from the ashes. This review was arranged to provide a comprehensive and precise description of MSCs' role in regulating tumorigenesis and to discuss the dark and the bright sides of cancer treatment strategies using MSCs. To gather the details about MSCs, we made an intensive literature review using keywords, including MSCs, tumor microenvironment, tumorigenesis, and targeted therapy. Through transferring cytokines, growth factors, and microRNAs, MSCs maintain the cancer stem cell population, increase angiogenesis, provide a facility for cancer metastasis, and shut down the anti-tumor activity of the immune system. Although MSCs progress tumorigenesis, there is a consensus that these cells could be used as a vehicle to transfer anti-cancer agents into the tumor milieu. This feature opened a new chapter in MSCs biology, this time from the therapeutic perspective. Although the data are not sufficient, the advent of new genetic engineering methods might make it possible to engage these cells as Trojan horses to eliminate the malignant population. So many years of investigation showed that MSCs are an important group of cells, residing in the TME, studying the function of which not only could add a delicate series of information to the process of tumorigenesis but also could revolutionize cancer treatment strategies.
Collapse
Affiliation(s)
- Mehdi Karimi-Shahri
- Department of Pathology, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Javid
- Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Sharbaf Mashhad
- Department of Medical Laboratory Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shaghayegh Yazdani
- Department of Medical Laboratory Sciences, Ilam Institute for Medical Sciences, Ilam, Iran
| | - Seyed Isaac Hashemy
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
33
|
Ghollasi M, Ghasembaglou S, Rahban D, Korani M, Motallebnezhad M, Asadi M, Zarredar H, Salimi A. Prospects for Manipulation of Mesenchymal Stem Cells in Tumor Therapy: Anti-Angiogenesis Property on the Spotlight. Int J Stem Cells 2021; 14:351-365. [PMID: 34456189 PMCID: PMC8611310 DOI: 10.15283/ijsc20146] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 06/01/2021] [Accepted: 06/16/2021] [Indexed: 11/10/2022] Open
Abstract
The interactions between the tumor microenvironment and the tumor cells confers a condition that accelerate or decelerate the development of tumor. Of these cells, mesenchymal stem cells (MSCs) have the potential to modulate the tumor cells. MSCs have been established with double functions, whereby contribute to a tumorigenic or anti-tumor setting. Clinical studies have indicated the potential of MSCs to be used as tool in treating the human cancer cells. One of the advantageous features of MSCs that make them as a well-suited tool for cancer therapy is the natural tumor-trophic migration potential. A key specification of the tumor development has been stablished to be angiogenesis. As a result, manipulation of angiogenesis has become an attractive approach for cancer therapy. This review article will seek to clarify the anti-angiogenesis strategy in modulating the MSCs to treat the tumor cells.
Collapse
Affiliation(s)
- Marzieh Ghollasi
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Shahram Ghasembaglou
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Dariush Rahban
- Department of Nanomedicine, School of Advanced Medical Technologies, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Korani
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Morteza Motallebnezhad
- Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Milad Asadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Basic Oncology, Ege University, Institute of Health Sciences, Izmir, Turkey
| | - Habib Zarredar
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Ali Salimi
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
34
|
Mesenchymal Stem Cells Can Both Enhance and Inhibit the Cellular Response to DNA Immunization by Genes of Nonstructural Proteins of the Hepatitis C Virus. Int J Mol Sci 2021. [DOI: 10.3390/ijms22158121
expr 825321411 + 858242883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Despite extensive research, there is still no vaccine against the hepatitis C virus (HCV). The aim of this study was to investigate whether MSCs can exhibit adjuvant properties during DNA vaccination against hepatitis C. We used the pcNS3-NS5B plasmid encoding five nonstructural HCV proteins and MSCs derived from mice bone marrow. Five groups of DBA mice were immunized with the plasmid and/or MSCs in a different order. Group 1 was injected with the plasmid twice at intervals of 3 weeks; Group 2 with the plasmid, and after 24 h with MSCs; Group 3 with MSCs followed by the plasmid the next day; Group 4 with only MSCs; and Group 5 with saline. When the MSCs were injected prior to DNA immunization, the cell immune response to HCV proteins assessed by the level of IFN-γ synthesis was markedly increased compared to DNA alone. In contrast, MSCs injected after DNA suppressed the immune response. Apparently, the high level of proinflammatory cytokines detected after DNA injection promotes the conversion of MSCs introduced later into the immunosuppressive MSC2. The low level of cytokines in mice before MSC administration promotes the high immunostimulatory activity of MSC1 in response to a DNA vaccine. Thus, when administered before DNA, MSCs are capable of exhibiting promising adjuvant properties.
Collapse
|
35
|
Masalova OV, Lesnova EI, Klimova RR, Ivanov AV, Kushch AA. Mesenchymal Stem Cells Can Both Enhance and Inhibit the Cellular Response to DNA Immunization by Genes of Nonstructural Proteins of the Hepatitis C Virus. Int J Mol Sci 2021; 22:8121. [PMID: 34360889 PMCID: PMC8347804 DOI: 10.3390/ijms22158121&set/a 880446214+990577611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 03/16/2023] Open
Abstract
Despite extensive research, there is still no vaccine against the hepatitis C virus (HCV). The aim of this study was to investigate whether MSCs can exhibit adjuvant properties during DNA vaccination against hepatitis C. We used the pcNS3-NS5B plasmid encoding five nonstructural HCV proteins and MSCs derived from mice bone marrow. Five groups of DBA mice were immunized with the plasmid and/or MSCs in a different order. Group 1 was injected with the plasmid twice at intervals of 3 weeks; Group 2 with the plasmid, and after 24 h with MSCs; Group 3 with MSCs followed by the plasmid the next day; Group 4 with only MSCs; and Group 5 with saline. When the MSCs were injected prior to DNA immunization, the cell immune response to HCV proteins assessed by the level of IFN-γ synthesis was markedly increased compared to DNA alone. In contrast, MSCs injected after DNA suppressed the immune response. Apparently, the high level of proinflammatory cytokines detected after DNA injection promotes the conversion of MSCs introduced later into the immunosuppressive MSC2. The low level of cytokines in mice before MSC administration promotes the high immunostimulatory activity of MSC1 in response to a DNA vaccine. Thus, when administered before DNA, MSCs are capable of exhibiting promising adjuvant properties.
Collapse
Affiliation(s)
- Olga V. Masalova
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.I.L.); (R.R.K.); (A.A.K.)
| | - Ekaterina I. Lesnova
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.I.L.); (R.R.K.); (A.A.K.)
| | - Regina R. Klimova
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.I.L.); (R.R.K.); (A.A.K.)
| | - Alexander V. Ivanov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Alla A. Kushch
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.I.L.); (R.R.K.); (A.A.K.)
| |
Collapse
|
36
|
Masalova OV, Lesnova EI, Klimova RR, Ivanov AV, Kushch AA. Mesenchymal Stem Cells Can Both Enhance and Inhibit the Cellular Response to DNA Immunization by Genes of Nonstructural Proteins of the Hepatitis C Virus. Int J Mol Sci 2021; 22:8121. [PMID: 34360889 PMCID: PMC8347804 DOI: 10.3390/ijms22158121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 12/21/2022] Open
Abstract
Despite extensive research, there is still no vaccine against the hepatitis C virus (HCV). The aim of this study was to investigate whether MSCs can exhibit adjuvant properties during DNA vaccination against hepatitis C. We used the pcNS3-NS5B plasmid encoding five nonstructural HCV proteins and MSCs derived from mice bone marrow. Five groups of DBA mice were immunized with the plasmid and/or MSCs in a different order. Group 1 was injected with the plasmid twice at intervals of 3 weeks; Group 2 with the plasmid, and after 24 h with MSCs; Group 3 with MSCs followed by the plasmid the next day; Group 4 with only MSCs; and Group 5 with saline. When the MSCs were injected prior to DNA immunization, the cell immune response to HCV proteins assessed by the level of IFN-γ synthesis was markedly increased compared to DNA alone. In contrast, MSCs injected after DNA suppressed the immune response. Apparently, the high level of proinflammatory cytokines detected after DNA injection promotes the conversion of MSCs introduced later into the immunosuppressive MSC2. The low level of cytokines in mice before MSC administration promotes the high immunostimulatory activity of MSC1 in response to a DNA vaccine. Thus, when administered before DNA, MSCs are capable of exhibiting promising adjuvant properties.
Collapse
Affiliation(s)
- Olga V. Masalova
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.I.L.); (R.R.K.); (A.A.K.)
| | - Ekaterina I. Lesnova
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.I.L.); (R.R.K.); (A.A.K.)
| | - Regina R. Klimova
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.I.L.); (R.R.K.); (A.A.K.)
| | - Alexander V. Ivanov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Alla A. Kushch
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.I.L.); (R.R.K.); (A.A.K.)
| |
Collapse
|
37
|
Bone marrow/bone pre-metastatic niche for breast cancer cells colonization: The role of mesenchymal stromal cells. Crit Rev Oncol Hematol 2021; 164:103416. [PMID: 34237436 DOI: 10.1016/j.critrevonc.2021.103416] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/17/2021] [Accepted: 07/04/2021] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is one of the most common oncological pathologies in women worldwide. While its early diagnosis has considerably improved, about 70 % of advanced patients develop bone metastases with a high mortality rate. Several authors demonstrated that primary breast cancer cells prepare their future metastatic niche -known as the pre-metastatic niche- to turn it into an "optimal soil" for colonization. The role of the different cellular components of the bone marrow/bone niche in bone metastasis has been well described. However, studying the changes that occur in this microenvironment before tumor cells arrival has become a novel research field. Therefore, the purpose of this review is to describe the current knowledge about the modulation of the normal bone marrow/bone niche by the primary breast tumor, in particular, highlighting the role of mesenchymal stem/stromal cells in transforming this soil into a pre-metastatic niche for breast cancer cells colonization.
Collapse
|
38
|
Ivolgin DA, Kudlay DA. Mesenchymal multipotent stromal cells and cancer safety: two sides of the same coin or a double-edged sword (review of foreign literature). RUSSIAN JOURNAL OF PEDIATRIC HEMATOLOGY AND ONCOLOGY 2021; 8:64-84. [DOI: 10.21682/2311-1267-2021-8-1-64-84] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Knowledge about the mechanisms of action of mesenchymal multipotent stromal cells (MSC) has undergone a significant evolution since their discovery. From the first attempts to use the remarkable properties of MSC in restoring the functions of organs and tissues, the most important question arose – how safe their use would be? One of the aspects of safety of the use of such biomaterial is tumorogenicity and oncogenicity. Numerous studies have shown that the mechanisms by which MSC realize their regenerative potential can, in principle, have a stimulating effect on tumor cells. This review presents specific mechanisms that have a potentially pro-tumor effect, which include the homing of MSC to the tumor site, support for replicative and proliferative signaling of both cancer cells and cancer stem cells, angiogenesis, and effects on the epithelial-mesenchymal transition. Along with pro-tumor mechanisms, the mechanisms of possible antitumor action are also described – direct suppression of tumor growth, loading and transportation of chemotherapeutic agents, oncolytic viruses, genetic modifications for targeting cancer, delivery of “suicide genes” to the tumor. Also, in conclusion, a small review of the current clinical trials of MSC as antitumor agents for malignant neoplasms of various localization (gastrointestinal tract, lungs, ovaries) is given.
Collapse
Affiliation(s)
- D. A. Ivolgin
- I.I. Mechnikov North-Western State Medical University, Ministry of Health of Russia
| | - D. A. Kudlay
- JSC “GENERIUM”;
I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia (Sechenov University);
National Research Center – Institute of Immunology Federal Medical-Biological Agency of Russia
| |
Collapse
|
39
|
Merckx G, Lo Monaco M, Lambrichts I, Himmelreich U, Bronckaers A, Wolfs E. Safety and Homing of Human Dental Pulp Stromal Cells in Head and Neck Cancer. Stem Cell Rev Rep 2021; 17:1619-1634. [PMID: 33822326 DOI: 10.1007/s12015-021-10159-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Head and neck cancer (HNC) is one of the most common cancers, associated with a huge mortality and morbidity. In order to improve patient outcomes, more efficient and targeted therapies are essential. Bone marrow-derived mesenchymal stromal cells (BM-MSCs) express tumour homing capacity, which could be exploited to target anti-cancer drug delivery to the tumour region and reduce adverse side-effects. Nevertheless, dental pulp stromal cells (DPSCs), an MSC-like population present in teeth, could offer important clinical benefits because of their easy isolation and superior proliferation compared to BM-MSCs. Therefore, we aimed to elucidate the tumour homing and safe usage of DPSCs to treat HNC. METHODS The in vivo survival as well as the effect of intratumourally administered DPSCs on tumour aggressiveness was tested in a HNC xenograft mouse model by using bioluminescence imaging (BLI), (immuno)histology and qRT-PCR. Furthermore, the in vitro and in vivo tumour homing capacity of DPSCs towards a HNC cell line were evaluated by a transwell migration assay and BLI, respectively. RESULTS Intratumourally injected DPSCs survived for at least two weeks in the tumour micro-environment and had no significant influence on tumour morphology, growth, angiogenesis and epithelial-to-mesenchymal transition. In addition, DPSCs migrated towards tumour cells in vitro, which could not be confirmed after their in vivo intravenous, intraperitoneal or peritumoural injection under the tested experimental conditions. CONCLUSIONS Our research suggests that intratumourally delivered DPSCs might be used as safe factories for the continuous delivery of anti-cancer drugs in HNC. Nevertheless, further optimization as well as efficacy studies are necessary to understand and improve in vivo tumour homing and determine the optimal experimental set-up of stem cell-based cancer therapies, including dosing and timing.
Collapse
Affiliation(s)
- Greet Merckx
- Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Group of Cardio & Organ Systems (COS), UHasselt - Hasselt University, Agoralaan, 3590, Diepenbeek, Belgium
| | - Melissa Lo Monaco
- Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Group of Cardio & Organ Systems (COS), UHasselt - Hasselt University, Agoralaan, 3590, Diepenbeek, Belgium.,Faculty of Sciences, Department of Veterinary Medicine, Integrated Veterinary Research Unit-Namur Research Institute for Life Science (IVRU-NARILIS), UNamur - University of Namur, Rue de Bruxelles, 5000, Namur, Belgium
| | - Ivo Lambrichts
- Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Group of Cardio & Organ Systems (COS), UHasselt - Hasselt University, Agoralaan, 3590, Diepenbeek, Belgium
| | - Uwe Himmelreich
- Department of Imaging and Pathology, Biomedical MRI Unit/MoSAIC, KU Leuven, Herestraat, 3000, Leuven, Belgium
| | - Annelies Bronckaers
- Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Group of Cardio & Organ Systems (COS), UHasselt - Hasselt University, Agoralaan, 3590, Diepenbeek, Belgium.
| | - Esther Wolfs
- Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Group of Cardio & Organ Systems (COS), UHasselt - Hasselt University, Agoralaan, 3590, Diepenbeek, Belgium
| |
Collapse
|
40
|
Abstract
Being the second leading cause of death globally, cancer has been a long-standing and rapidly evolving focus of biomedical research and practice in the world. A tremendous effort has been made to understand the origin of cancer cells, the formation of cancerous tissues, and the mechanism by which they spread and relapse, but the disease still remains mysterious. Here, we made an attempt to scrutinize evidences that indicate the role of stem cells in tumorigenesis and metastasis, and cancer relapse. We also looked into the influence of cancers on stem cells, which in turn represent a major constituent of tumor microenvironment. Based on current understandings of the properties of (cancer) stem cells and their relation to cancers, we can foresee that novel therapeutic approaches would become the next wave of cancer treatment.
Collapse
Affiliation(s)
- Wen Yin
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Sichuan 610041, China
| | - Jialing Wang
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Sichuan 610041, China
| | - Linling Jiang
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Sichuan 610041, China
| | - Y James Kang
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Sichuan 610041, China.,Memphis Institute of Regenerative Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
41
|
Piccotti F, Rybinska I, Scoccia E, Morasso C, Ricciardi A, Signati L, Triulzi T, Corsi F, Truffi M. Lipofilling in Breast Oncological Surgery: A Safe Opportunity or Risk for Cancer Recurrence? Int J Mol Sci 2021; 22:ijms22073737. [PMID: 33916703 PMCID: PMC8038405 DOI: 10.3390/ijms22073737] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 02/08/2023] Open
Abstract
Lipofilling (LF) is a largely employed technique in reconstructive and esthetic breast surgery. Over the years, it has demonstrated to be extremely useful for treatment of soft tissue defects after demolitive or conservative breast cancer surgery and different procedures have been developed to improve the survival of transplanted fat graft. The regenerative potential of LF is attributed to the multipotent stem cells found in large quantity in adipose tissue. However, a growing body of pre-clinical evidence shows that adipocytes and adipose-derived stromal cells may have pro-tumorigenic potential. Despite no clear indication from clinical studies has demonstrated an increased risk of cancer recurrence upon LF, these observations challenge the oncologic safety of the procedure. This review aims to provide an updated overview of both the clinical and the pre-clinical indications to the suitability and safety of LF in breast oncological surgery. Cellular and molecular players in the crosstalk between adipose tissue and cancer are described, and heterogeneous contradictory results are discussed, highlighting that important issues still remain to be solved to get a clear understanding of LF safety in breast cancer patients.
Collapse
Affiliation(s)
- Francesca Piccotti
- Laboratorio di Nanomedicina ed Imaging Molecolare, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (F.P.); (C.M.); (A.R.)
| | - Ilona Rybinska
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (I.R.); (T.T.)
| | - Elisabetta Scoccia
- Breast Unit, Surgery Department, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (E.S.); (F.C.)
| | - Carlo Morasso
- Laboratorio di Nanomedicina ed Imaging Molecolare, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (F.P.); (C.M.); (A.R.)
| | - Alessandra Ricciardi
- Laboratorio di Nanomedicina ed Imaging Molecolare, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (F.P.); (C.M.); (A.R.)
| | - Lorena Signati
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università Degli Studi di Milano, 20157 Milano, Italy;
| | - Tiziana Triulzi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (I.R.); (T.T.)
| | - Fabio Corsi
- Breast Unit, Surgery Department, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (E.S.); (F.C.)
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università Degli Studi di Milano, 20157 Milano, Italy;
| | - Marta Truffi
- Laboratorio di Nanomedicina ed Imaging Molecolare, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (F.P.); (C.M.); (A.R.)
- Correspondence: ; Tel.: +39-0382-592219
| |
Collapse
|
42
|
The Potential of Mesenchymal Stromal Cells in Neuroblastoma Therapy for Delivery of Anti-Cancer Agents and Hematopoietic Recovery. J Pers Med 2021; 11:jpm11030161. [PMID: 33668854 PMCID: PMC7996318 DOI: 10.3390/jpm11030161] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/19/2021] [Accepted: 02/21/2021] [Indexed: 02/07/2023] Open
Abstract
Neuroblastoma is one of the most common pediatric cancers and a major cause of cancer-related death in infancy. Conventional therapies including high-dose chemotherapy, stem cell transplantation, and immunotherapy approach a limit in the treatment of high-risk neuroblastoma and prevention of relapse. In the last two decades, research unraveled a potential use of mesenchymal stromal cells in tumor therapy, as tumor-selective delivery vehicles for therapeutic compounds and oncolytic viruses and by means of supporting hematopoietic stem cell transplantation. Based on pre-clinical and clinical advances in neuroblastoma and other malignancies, we assess both the strong potential and the associated risks of using mesenchymal stromal cells in the therapy for neuroblastoma. Furthermore, we examine feasibility and safety aspects and discuss future directions for harnessing the advantageous properties of mesenchymal stromal cells for the advancement of therapy success.
Collapse
|
43
|
Moreno R. Mesenchymal stem cells and oncolytic viruses: joining forces against cancer. J Immunother Cancer 2021; 9:e001684. [PMID: 33558278 PMCID: PMC7871674 DOI: 10.1136/jitc-2020-001684] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2020] [Indexed: 12/13/2022] Open
Abstract
The development of oncolytic viruses (OVs) has increased significantly in the past 20 years, with many candidates entering clinical trials and three of them receiving approval for some indications. Recently, OVs have also gathered interest as candidates to use in combination with immunotherapies for cancer due to their immunogenic properties, which include immunogenic cell death and the possibility to carry therapeutic transgenes in their genomes. OVs transform non-immunogenic 'cold' tumors into inflamed immunogenic 'hot' tumors, where immunotherapies show the highest efficacy. However, in monotherapy or in combination with immunotherapy, OVs face numerous challenges that limit their successful application, in particular upon systemic administration, such as liver sequestration, neutralizing interactions in blood, physical barriers to infection, and fast clearance by the immune system. In this regard, the use of mesenchymal stem cells (MSCs) as cells carrier for OV delivery addresses many of these obstacles acting as virus carriers and factories, expressing additional transgenes, and modulating the immune system. Here, I review the current progress of OVs-loaded MSCs in cancer, focusing on their interaction with the immune system, and discuss new strategies to improve their therapeutic efficacy.
Collapse
Affiliation(s)
- Rafael Moreno
- Virotherapy and immunotherapy group, ProCURE Program, Catalan Institute of Oncology - ICO, L'Hospitalet de Llobregat, Spain
- Cancer Virotherapy group, Oncobell Program, Institutd'Investigació Biomèdica de Bellvitge - IDIBELL, L'Hospitalet de Llobregat, Spain
| |
Collapse
|
44
|
Gholizadeh-Ghaleh Aziz S, Alipour S, Ranjbarvan P, Azari A, Babaei G, Golchin A. Critical roles of TLRs on the polarization of mesenchymal stem cells for cell therapy of viral infections: a notice for COVID-19 treatment. COMPARATIVE CLINICAL PATHOLOGY 2021; 30:119-128. [PMID: 33551714 PMCID: PMC7846495 DOI: 10.1007/s00580-021-03209-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 01/24/2021] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cells (MSCs), as one of the leading cell-based therapy, have provided a strong link between clinical investigation and basic research. MSCs have been successfully employed in treating graft versus host disease (GvHD), autoimmune disease, and several other diseases, particularly with high immune activity. Recently, MSCs have attracted attention to treating untreatable viral infections such as severe coronavirus disease 2019 (COVID-19). Given that the Toll-like receptors (TLRs) are directly able to detect internal and external hazard signals, and their stimulation has an intense effect on the ability to grow, differentiate, migrate, and maintain MSCs, it seems stimulation of these receptors can have a direct impact on the interaction of MSCs and immune cells, altering the ability to modify immune system responses. Hence, this mini-review focused on TLRs' critical roles in the polarization of MSCs for developing MSC-based therapy in viral infections. Consequently, according to the literature review, a polarization process, mediated by TLRs concerning anti-inflammatory and proinflammatory phenotype, may be considered for MSC-therapy against viral infections.
Collapse
Affiliation(s)
- Shiva Gholizadeh-Ghaleh Aziz
- Department of Clinical Biochemistry and Applied Cell Sciences, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Shahriar Alipour
- Department of Clinical Biochemistry and Applied Cell Sciences, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Parviz Ranjbarvan
- Department of Clinical Biochemistry and Applied Cell Sciences, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Arezo Azari
- Department of Tissue engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ghader Babaei
- Department of Clinical Biochemistry and Applied Cell Sciences, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Ali Golchin
- Department of Clinical Biochemistry and Applied Cell Sciences, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
45
|
Laplace-Builhé B, Bahraoui S, Jorgensen C, Djouad F. From the Basis of Epimorphic Regeneration to Enhanced Regenerative Therapies. Front Cell Dev Biol 2021; 8:605120. [PMID: 33585444 PMCID: PMC7873919 DOI: 10.3389/fcell.2020.605120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 12/14/2020] [Indexed: 01/01/2023] Open
Abstract
Current cell-based therapies to treat degenerative diseases such as osteoarthritis (OA) fail to offer long-term beneficial effects. The therapeutic effects provided by mesenchymal stem cell (MSC) injection, characterized by reduced pain and an improved functional activity in patients with knee OA, are reported at short-term follow-up since the improved outcomes plateau or, even worse, decline several months after MSC administration. This review tackles the limitations of MSC-based therapy for degenerative diseases and highlights the lessons learned from regenerative species to comprehend the coordination of molecular and cellular events critical for complex regeneration processes. We discuss how MSC injection generates a positive cascade of events resulting in a long-lasting systemic immune regulation with limited beneficial effects on tissue regeneration while in regenerative species fine-tuned inflammation is required for progenitor cell proliferation, differentiation, and regeneration. Finally, we stress the direct or indirect involvement of neural crest derived cells (NCC) in most if not all adult regenerative models studied so far. This review underlines the regenerative potential of NCC and the limitations of MSC-based therapy to open new avenues for the treatment of degenerative diseases such as OA.
Collapse
Affiliation(s)
| | | | - Christian Jorgensen
- IRMB, Univ Montpellier, INSERM, Montpellier, France.,CHU Montpellier, Montpellier, France
| | | |
Collapse
|
46
|
Kouroupis D, Willman MA, Best TM, Kaplan LD, Correa D. Infrapatellar fat pad-derived mesenchymal stem cell-based spheroids enhance their therapeutic efficacy to reverse synovitis and fat pad fibrosis. Stem Cell Res Ther 2021; 12:44. [PMID: 33413649 PMCID: PMC7792122 DOI: 10.1186/s13287-020-02107-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/15/2020] [Indexed: 12/13/2022] Open
Abstract
Background To investigate the in vitro and in vivo anti-inflammatory/anti-fibrotic capacity of IFP-MSC manufactured as 3D spheroids. Our hypothesis is that IFP-MSC do not require prior cell priming to acquire a robust immunomodulatory phenotype in vitro in order to efficiently reverse synovitis and IFP fibrosis, and secondarily delay articular cartilage damage in vivo. Methods Human IFP-MSC immunophenotype, tripotentiality, and transcriptional profiles were assessed in 3D settings. Multiplex secretomes were assessed in IFP-MSC spheroids [Crude (non-immunoselected), CD146+ or CD146− immunoselected cells] and compared with 2D cultures with and without prior inflammatory/fibrotic cell priming. Functionally, IFP-MSC spheroids were assessed for their immunopotency on human PBMC proliferation and their effect on stimulated synoviocytes with inflammation and fibrotic cues. The anti-inflammatory and anti-fibrotic spheroid properties were further evaluated in vivo in a rat model of acute synovitis/fat pad fibrosis. Results Spheroids enhanced IFP-MSC phenotypic, transcriptional, and secretory immunomodulatory profiles compared to 2D cultures. Further, CD146+ IFP-MSC spheroids showed enhanced secretory and transcriptional profiles; however, these attributes were not reflected in a superior capacity to suppress activated PBMC. This suggests that 3D culturing settings are sufficient to induce an enhanced immunomodulatory phenotype in both Crude and CD146-immunoselected IFP-MSC. Crude IFP-MSC spheroids modulated the molecular response of synoviocytes previously exposed to inflammatory cues. Therapeutically, IFP-MSC spheroids retained substance P degradation potential in vivo, while effectively inducing resolution of inflammation/fibrosis of the synovium and fat pad. Furthermore, their presence resulted in arrest of articular cartilage degradation in a rat model of progressive synovitis and fat pad fibrosis. Conclusions 3D spheroids confer IFP-MSC a reproducible and enhanced immunomodulatory effect in vitro and in vivo, circumventing the requirement of non-compliant cell priming or selection before administration and thereby streamlining cell products manufacturing protocols.
Collapse
Affiliation(s)
- Dimitrios Kouroupis
- Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami, Miller School of Medicine, 1450 NW 10th Ave (3014), Miami, FL, 33136, USA.,Diabetes Research Institute & Cell Transplantation Center, University of Miami, Miller School of Medicine, 1450 NW 10th Ave (3014), Miami, FL, 33136, USA
| | - Melissa A Willman
- Diabetes Research Institute & Cell Transplantation Center, University of Miami, Miller School of Medicine, 1450 NW 10th Ave (3014), Miami, FL, 33136, USA
| | - Thomas M Best
- Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami, Miller School of Medicine, 1450 NW 10th Ave (3014), Miami, FL, 33136, USA
| | - Lee D Kaplan
- Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami, Miller School of Medicine, 1450 NW 10th Ave (3014), Miami, FL, 33136, USA
| | - Diego Correa
- Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami, Miller School of Medicine, 1450 NW 10th Ave (3014), Miami, FL, 33136, USA. .,Diabetes Research Institute & Cell Transplantation Center, University of Miami, Miller School of Medicine, 1450 NW 10th Ave (3014), Miami, FL, 33136, USA.
| |
Collapse
|
47
|
Stevens HY, Bowles AC, Yeago C, Roy K. Molecular Crosstalk Between Macrophages and Mesenchymal Stromal Cells. Front Cell Dev Biol 2020; 8:600160. [PMID: 33363157 PMCID: PMC7755599 DOI: 10.3389/fcell.2020.600160] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/05/2020] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have been widely investigated for regenerative medicine applications, from treating various inflammatory diseases as a cell therapy to generating engineered tissue constructs. Numerous studies have evaluated the potential effects of MSCs following therapeutic administration. By responding to their surrounding microenvironment, MSCs may mediate immunomodulatory effects through various mechanisms that directly (i.e., contact-dependent) or indirectly (i.e., paracrine activity) alter the physiology of endogenous cells in various disease pathologies. More specifically, a pivotal crosstalk between MSCs and tissue-resident macrophages and monocytes (TMφ) has been elucidated using in vitro and in vivo preclinical studies. An improved understanding of this crosstalk could help elucidate potential mechanisms of action (MOAs) of therapeutically administered MSCs. TMφ, by nature of their remarkable functional plasticity and prevalence within the body, are uniquely positioned as critical modulators of the immune system - not only in maintaining homeostasis but also during pathogenesis. This has prompted further exploration into the cellular and molecular alterations to TMφ mediated by MSCs. In vitro assays and in vivo preclinical trials have identified key interactions mediated by MSCs that polarize the responses of TMφ from a pro-inflammatory (i.e., classical activation) to a more anti-inflammatory/reparative (i.e., alternative activation) phenotype and function. In this review, we describe physiological and pathological TMφ functions in response to various stimuli and discuss the evidence that suggest specific mechanisms through which MSCs may modulate TMφ phenotypes and functions, including paracrine interactions (e.g., secretome and extracellular vesicles), nanotube-mediated intercellular exchange, bioenergetics, and engulfment by macrophages. Continued efforts to elucidate this pivotal crosstalk may offer an improved understanding of the immunomodulatory capacity of MSCs and inform the development and testing of potential MOAs to support the therapeutic use of MSCs and MSC-derived products in various diseases.
Collapse
Affiliation(s)
- Hazel Y. Stevens
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
| | - Annie C. Bowles
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
| | - Carolyn Yeago
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
- NSF Engineering Research Center (ERC) for Cell Manufacturing Technologies (CMaT), Georgia Institute of Technology, Atlanta, GA, United States
| | - Krishnendu Roy
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
- NSF Engineering Research Center (ERC) for Cell Manufacturing Technologies (CMaT), Georgia Institute of Technology, Atlanta, GA, United States
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
- Center for ImmunoEngineering, Georgia Institute of Technology, Atlanta, GA, United States
| |
Collapse
|
48
|
Dagnino APA, Chagastelles PC, Medeiros RP, Estrázulas M, Kist LW, Bogo MR, Weber JBB, Campos MM, Silva JB. Neural Regenerative Potential of Stem Cells Derived from the Tooth Apical Papilla. Stem Cells Dev 2020; 29:1479-1496. [PMID: 32988295 DOI: 10.1089/scd.2020.0121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The regenerative effects of stem cells derived from dental tissues have been previously investigated. This study assessed the potential of human tooth stem cells from apical papilla (SCAP) on nerve regeneration. The SCAP collected from nine individuals were characterized and polarized by exposure to interferon-γ (IFN-γ). IFN-γ increased kynurenine and interleukin-6 (IL-6) production by SCAP, without affecting the cell viability. IFN-γ-primed SCAP exhibited a decrease of brain-derived neurotrophic factor (BDNF) mRNA levels, followed by an upregulation of glial cell-derived neurotrophic factor mRNA. Ex vivo, the co-culture of SCAP with neurons isolated from the rat dorsal root ganglion induced neurite outgrowth, accompanied by increased BDNF secretion, irrespective of IFN-γ priming. In vivo, the local application of SCAP reduced the mechanical and thermal hypersensitivity in Wistar rats that had been submitted to sciatic chronic constriction injury. The SCAP also reduced the pain scores, according to the evaluation of the Grimace scale, partially restoring the myelin damage and BDNF immunopositivity secondary to nerve lesion. Altogether, our results provide novel evidence about the regenerative effects of human SCAP, indicating their potential to handle nerve injury-related complications.
Collapse
Affiliation(s)
- Ana Paula Aquistapase Dagnino
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Centro de Pesquisa em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Pedro Cesar Chagastelles
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Renata Priscila Medeiros
- Centro de Pesquisa em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Programa de Pós-Graduação em Odontologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Marina Estrázulas
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Centro de Pesquisa em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Luiza Wilges Kist
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Laboratório de Biologia Genômica e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Maurício Reis Bogo
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Laboratório de Biologia Genômica e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - João Batista Blessmann Weber
- Programa de Pós-Graduação em Odontologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Maria Martha Campos
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Centro de Pesquisa em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Programa de Pós-Graduação em Odontologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Jefferson Braga Silva
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| |
Collapse
|
49
|
Zheng P, Li W. Crosstalk Between Mesenchymal Stromal Cells and Tumor-Associated Macrophages in Gastric Cancer. Front Oncol 2020; 10:571516. [PMID: 33163402 PMCID: PMC7581781 DOI: 10.3389/fonc.2020.571516] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 09/08/2020] [Indexed: 12/12/2022] Open
Abstract
Tumor microenvironment (TME) consisting of distinct cell types including stromal cells and immune cells has recently emerged as a pivotal player in tumor development and progression. Mesenchymal stromal cells (MSCs) and tumor-associated macrophages (TAMs) are two representative cells in the TME with plastic properties. This review will focus on the evolution of phenotypes and functions of either MSCs or TAMs, which is “educated” by the TME, as well as interactions between MSCs and TAMs contributing to the distinct stages of tumor biology in gastric cancer. MSCs exert immunoregulatory effects on macrophages and polarize them toward M2-like TAMs, via cell–cell contact and paracrine or extracellular vesicle (EV) transfer mechanism. In turn, M2-TAMs modulate the transition of “naive” MSCs into tumor-derived MSCs, which possess a more potent pro-tumor role than the parent. Moreover, the cross talk between MSCs and TAMs could contribute to cancer biology by inducing the EMT process, metastasis, immune invasion, and immunotherapy resistance in cancer cells. However, molecular mechanisms underlying interactions between MSCs and TAMs in gastric cancer progression need to be thoroughly elucidated, which may provide attractive targets for making promising novel strategies for gastric cancer therapy.
Collapse
Affiliation(s)
- Ping Zheng
- Department of Laboratory Medicine, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Wei Li
- Center of Research Laboratory, The First People's Hospital of Lianyungang, Lianyungang, China
| |
Collapse
|
50
|
Three-Dimensional Culture System of Cancer Cells Combined with Biomaterials for Drug Screening. Cancers (Basel) 2020; 12:cancers12102754. [PMID: 32987868 PMCID: PMC7601447 DOI: 10.3390/cancers12102754] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/17/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary For the research and development of drug discovery, it is of prime importance to construct the three-dimensional (3D) tissue models in vitro. To this end, the enhancement design of cell function and activity by making use of biomaterials is essential. In this review, 3D culture systems of cancer cells combined with several biomaterials for anticancer drug screening are introduced. Abstract Anticancer drug screening is one of the most important research and development processes to develop new drugs for cancer treatment. However, there is a problem resulting in gaps between the in vitro drug screening and preclinical or clinical study. This is mainly because the condition of cancer cell culture is quite different from that in vivo. As a trial to mimic the in vivo cancer environment, there has been some research on a three-dimensional (3D) culture system by making use of biomaterials. The 3D culture technologies enable us to give cancer cells an in vitro environment close to the in vivo condition. Cancer cells modified to replicate the in vivo cancer environment will promote the biological research or drug discovery of cancers. This review introduces the in vitro research of 3D cell culture systems with biomaterials in addition to a brief summary of the cancer environment.
Collapse
|