1
|
Use of Brain-Derived Stem/Progenitor Cells and Derived Extracellular Vesicles to Repair Damaged Neural Tissues: Lessons Learned from Connective Tissue Repair Regarding Variables Limiting Progress and Approaches to Overcome Limitations. Int J Mol Sci 2023; 24:ijms24043370. [PMID: 36834779 PMCID: PMC9958575 DOI: 10.3390/ijms24043370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/27/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Pluripotent neural stem or progenitor cells (NSC/NPC) have been reported in the brains of adult preclinical models for decades, as have mesenchymal stem/stromal cells (MSC) been reported in a variety of tissues from adults. Based on their in vitro capabilities, these cell types have been used extensively in attempts to repair/regenerate brain and connective tissues, respectively. In addition, MSC have also been used in attempts to repair compromised brain centres. However, success in treating chronic neural degenerative conditions such as Alzheimer's disease, Parkinson's disease, and others with NSC/NPC has been limited, as have the use of MSC in the treatment of chronic osteoarthritis, a condition affecting millions of individuals. However, connective tissues are likely less complex than neural tissues regarding cell organization and regulatory integration, but some insights have been gleaned from the studies regarding connective tissue healing with MSC that may inform studies attempting to initiate repair and regeneration of neural tissues compromised acutely or chronically by trauma or disease. This review will discuss the similarities and differences in the applications of NSC/NPC and MSC, where some lessons have been learned, and potential approaches that could be used going forward to enhance progress in the application of cellular therapy to facilitate repair and regeneration of complex structures in the brain. In particular, variables that may need to be controlled to enhance success are discussed, as are different approaches such as the use of extracellular vesicles from stem/progenitor cells that could be used to stimulate endogenous cells to repair the tissues rather than consider cell replacement as the primary option. Caveats to all these efforts relate to whether cellular repair initiatives will have long-term success if the initiators for neural diseases are not controlled, and whether such cellular initiatives will have long-term success in a subset of patients if the neural diseases are heterogeneous and have multiple etiologies.
Collapse
|
2
|
Do ‘nature’ mediate stem cells empowerment in women? Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2022.110963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
3
|
Välimäki NN, Bakreen A, Häkli S, Dhungana H, Keuters MH, Dunlop Y, Koskuvi M, Keksa-Goldsteine V, Oksanen M, Jäntti H, Lehtonen Š, Malm T, Koistinaho J, Jolkkonen J. Astrocyte Progenitors Derived From Patients With Alzheimer Disease Do Not Impair Stroke Recovery in Mice. Stroke 2022; 53:3192-3201. [DOI: 10.1161/strokeaha.122.039700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND:
Species-specific differences in astrocytes and their Alzheimer disease-associated pathology may influence cellular responses to other insults. Herein, human glial chimeric mice were generated to evaluate how Alzheimer disease predisposing genetic background in human astrocytes contributes to behavioral outcome and brain pathology after cortical photothrombotic ischemia.
METHODS:
Neonatal (P0) immunodeficient mice of both sexes were transplanted with induced pluripotent stem cell–derived astrocyte progenitors from Alzheimer disease patients carrying
PSEN1
exon 9 deletion (
P
SEN1
Δ
E
9), with isogenic controls, with cells from a healthy donor, or with mouse astrocytes or vehicle. After 14 months, a photothrombotic lesion was produced with Rose Bengal in the motor cortex. Behavior was assessed before ischemia and 1 and 4 weeks after the induction of stroke, followed by tissue perfusion for histology.
RESULTS:
Open field, cylinder, and grid-walking tests showed a persistent locomotor and sensorimotor impairment after ischemia and female mice had larger infarct sizes; yet, these were not affected by astrocytes with
P
SEN1
Δ
E
9 background. Staining for human nuclear antigen confirmed that human cells successfully engrafted throughout the mouse brain. However, only a small number of human cells were positive for astrocytic marker GFAP (glial fibrillary acidic protein), mostly located in the corpus callosum and retaining complex human-specific morphology with longer processes compared with host counterparts. While host astrocytes formed the glial scar, human astrocytes were scattered in small numbers close to the lesion boundary. Aβ (beta-amyloid) deposits were not present in
P
SEN1
ΔE
9 astrocyte-transplanted mice.
CONCLUSIONS:
Transplanted human cells survived and distributed widely in the host brain but had no impact on severity of ischemic damage after cortical photothrombosis in chimeric mice. Only a small number of transplanted human astrocytes acquired GFAP-positive glial phenotype or migrated toward the ischemic lesion forming glial scar.
P
SEN1
ΔE
9 astrocytes did not impair behavioral recovery after experimental stroke.
Collapse
Affiliation(s)
- Nelli-Noora Välimäki
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
| | - Abdulhameed Bakreen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
| | - Sara Häkli
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
| | - Hiramani Dhungana
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
- Neuroscience Center, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Finland (H.D., M.H.K., M.K., Š.L., J.K.)
| | - Meike H. Keuters
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
- Neuroscience Center, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Finland (H.D., M.H.K., M.K., Š.L., J.K.)
| | - Yannick Dunlop
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
| | - Marja Koskuvi
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
- Neuroscience Center, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Finland (H.D., M.H.K., M.K., Š.L., J.K.)
| | - Velta Keksa-Goldsteine
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
| | - Minna Oksanen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
| | - Henna Jäntti
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
| | - Šárka Lehtonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
- Neuroscience Center, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Finland (H.D., M.H.K., M.K., Š.L., J.K.)
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
| | - Jari Koistinaho
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
- Neuroscience Center, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Finland (H.D., M.H.K., M.K., Š.L., J.K.)
| | - Jukka Jolkkonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
| |
Collapse
|
4
|
Bedell ML, Torres AL, Hogan KJ, Wang Z, Wang B, Melchiorri AJ, Grande-Allen KJ, Mikos AG. Human gelatin-based composite hydrogels for osteochondral tissue engineering and their adaptation into bioinks for extrusion, inkjet, and digital light processing bioprinting. Biofabrication 2022; 14:10.1088/1758-5090/ac8768. [PMID: 35931060 PMCID: PMC9633045 DOI: 10.1088/1758-5090/ac8768] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 08/04/2022] [Indexed: 11/11/2022]
Abstract
The investigation of novel hydrogel systems allows for the study of relationships between biomaterials, cells, and other factors within osteochondral tissue engineering. Three-dimensional (3D) printing is a popular research method that can allow for further interrogation of these questions via the fabrication of 3D hydrogel environments that mimic tissue-specific, complex architectures. However, the adaptation of promising hydrogel biomaterial systems into 3D-printable bioinks remains a challenge. Here, we delineated an approach to that process. First, we characterized a novel methacryloylated gelatin composite hydrogel system and assessed how calcium phosphate and glycosaminoglycan additives upregulated bone- and cartilage-like matrix deposition and certain genetic markers of differentiation within human mesenchymal stem cells (hMSCs), such as RUNX2 and SOX9. Then, new assays were developed and utilized to study the effects of xanthan gum and nanofibrillated cellulose, which allowed for cohesive fiber deposition, reliable droplet formation, and non-fracturing digital light processing (DLP)-printed constructs within extrusion, inkjet, and DLP techniques, respectively. Finally, these bioinks were used to 3D print constructs containing viable encapsulated hMSCs over a 7 d period, where DLP printed constructs facilitated the highest observed increase in cell number over 7 d (∼2.4×). The results presented here describe the promotion of osteochondral phenotypes via these novel composite hydrogel formulations, establish their ability to bioprint viable, cell-encapsulating constructs using three different 3D printing methods on multiple bioprinters, and document how a library of modular bioink additives affected those physicochemical properties important to printability.
Collapse
Affiliation(s)
| | | | - Katie J. Hogan
- Department of Bioengineering, Rice University, Houston, TX
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX
| | - Ziwen Wang
- Department of Bioengineering, Rice University, Houston, TX
| | - Bonnie Wang
- Department of Bioengineering, Rice University, Houston, TX
| | | | | | - Antonios G. Mikos
- Department of Bioengineering, Rice University, Houston, TX
- NIBIB/NIH Center for Engineering Complex Tissues, USA
| |
Collapse
|
5
|
Knewtson KE, Ohl NR, Robinson JL. Estrogen Signaling Dictates Musculoskeletal Stem Cell Behavior: Sex Differences in Tissue Repair. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:789-812. [PMID: 34409868 PMCID: PMC9419932 DOI: 10.1089/ten.teb.2021.0094] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Sexual dimorphisms in humans and other species exist in visually evident features such as body size and less apparent characteristics, including disease prevalence. Current research is adding to a growing understanding of sex differences in stem cell function and response to external stimuli, including sex hormones such as estrogens. These differences are proving significant and directly impact both the understanding of stem cell processes in tissue repair and the clinical implementation of stem cell therapies. Adult stem cells of the musculoskeletal system, including those used for development and repair of muscle, bone, cartilage, fibrocartilage, ligaments, and tendons, are no exception. Both in vitro and in vivo studies have found differences in stem cell number, proliferative and differentiation capabilities, and response to estrogen treatment between males and females of many species. Maintaining the stemness and reducing senescence of adult stem cells is an important topic with implications in regenerative therapy and aging. As such, this review discusses the effect of estrogens on musculoskeletal system stem cell response in multiple species and highlights the research gaps that still need to be addressed. The following evidence from investigations of sex-related phenotypes in adult progenitor and stem cells are pieces to the big puzzle of sex-related effects on aging and disease and critical information for both fundamental tissue repair and regeneration studies and safe and effective clinical use of stem cells. Impact Statement This review summarizes current knowledge of sex differences in and the effects of estrogen treatment on musculoskeletal stem cells in the context of tissue engineering. Specifically, it highlights the impact of sex on musculoskeletal stem cell function and ability to regenerate tissue. Furthermore, it discusses the varying effects of estrogen on stem cell properties, including proliferation and differentiation, important to tissue engineering. This review aims to highlight the potential impact of estrogens and the importance of performing sex comparative studies in the field of tissue engineering.
Collapse
Affiliation(s)
- Kelsey E. Knewtson
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas, USA
| | - Nathan R. Ohl
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas, USA
| | - Jennifer L. Robinson
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas, USA
- Bioengineering Graduate Program, University of Kansas, Lawrence, Kansas, USA
- Address correspondence to: Jennifer L. Robinson, PhD, Department of Chemical and Petroleum Engineering, The University of Kansas, 1530 West 15th Street Room 4132, Lawrence, KS 66045, USA
| |
Collapse
|
6
|
Calcat-i-Cervera S, Sanz-Nogués C, O'Brien T. When Origin Matters: Properties of Mesenchymal Stromal Cells From Different Sources for Clinical Translation in Kidney Disease. Front Med (Lausanne) 2021; 8:728496. [PMID: 34616756 PMCID: PMC8488400 DOI: 10.3389/fmed.2021.728496] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/19/2021] [Indexed: 12/14/2022] Open
Abstract
Advanced therapy medicinal products (ATMPs) offer new prospects to improve the treatment of conditions with unmet medical needs. Kidney diseases are a current major health concern with an increasing global prevalence. Chronic renal failure appears after many years of impairment, which opens a temporary window to apply novel therapeutic approaches to delay or halt disease progression. The immunomodulatory, anti-inflammatory, and pro-regenerative properties of mesenchymal stromal cells (MSCs) have sparked interest for their use in cell-based regenerative therapies. Currently, several early-phase clinical trials have been completed and many are ongoing to explore MSC safety and efficacy in a wide range of nephropathies. However, one of the current roadblocks to the clinical translation of MSC therapies relates to the lack of standardization and harmonization of MSC manufacturing protocols, which currently hinders inter-study comparability. Studies have shown that cell culture processing variables can have significant effects on MSC phenotype and functionality, and these are highly variable across laboratories. In addition, heterogeneity within MSC populations is another obstacle. Furthermore, MSCs may be isolated from several sources which adds another variable to the comparative assessment of outcomes. There is now a growing body of literature highlighting unique and distinctive properties of MSCs according to the tissue origin, and that characteristics such as donor, age, sex and underlying medical conditions may alter the therapeutic effect of MSCs. These variables must be taken into consideration when developing a cell therapy product. Having an optimal scale-up strategy for MSC manufacturing is critical for ensuring product quality while minimizing costs and time of production, as well as avoiding potential risks. Ideally, optimal scale-up strategies must be carefully considered and identified during the early stages of development, as making changes later in the bioprocess workflow will require re-optimization and validation, which may have a significant long-term impact on the cost of the therapy. This article provides a summary of important cell culture processing variables to consider in the scale-up of MSC manufacturing as well as giving a comprehensive review of tissue of origin-specific biological characteristics of MSCs and their use in current clinical trials in a range of renal pathologies.
Collapse
Affiliation(s)
| | | | - Timothy O'Brien
- Regenerative Medicine Institute (REMEDI), CÚRAM, Biomedical Science Building, National University of Ireland, Galway, Ireland
| |
Collapse
|
7
|
Human Umbilical Cord: Information Mine in Sex-Specific Medicine. Life (Basel) 2021; 11:life11010052. [PMID: 33451112 PMCID: PMC7828611 DOI: 10.3390/life11010052] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/08/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Biological differences between sexes should be considered in all stages of research, as sexual dimorphism starts in utero leading to sex-specific fetal programming. In numerous biomedical fields, there is still a lack of stratification by sex despite primary cultured cells retaining memory of the sex and of the donor. The sex of donors in biological research must be known because variations in cells and cellular components can be used as endpoints, biomarkers and/or targets of pharmacological studies. This selective review focuses on the current findings regarding sex differences observed in the umbilical cord, a widely used source of research samples, both in the blood and in the circulating cells, as well as in the different cellular models obtainable from it. Moreover, an overview on sex differences in fetal programming is reported. As it emerges that the sex variable is still often forgotten in experimental models, we suggest that it should be mandatory to adopt sex-oriented research, because only awareness of these issues can lead to innovative research.
Collapse
|
8
|
Florea V, Rieger AC, Natsumeda M, Tompkins BA, Banerjee MN, Schulman IH, Premer C, Khan A, Valasaki K, Heidecker B, Mantero A, Balkan W, Mitrani RD, Hare JM. The impact of patient sex on the response to intramyocardial mesenchymal stem cell administration in patients with non-ischaemic dilated cardiomyopathy. Cardiovasc Res 2020; 116:2131-2141. [PMID: 32053144 PMCID: PMC7584465 DOI: 10.1093/cvr/cvaa004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 10/31/2019] [Accepted: 02/05/2020] [Indexed: 12/11/2022] Open
Abstract
AIMS Sex differences impact the occurrence, presentation, prognosis, and response to therapy in heart disease. Particularly, the phenotypic presentation of patients with non-ischaemic dilated cardiomyopathy (NIDCM) differs between men and women. However, whether the response to mesenchymal stem cell (MSC) therapy is influenced by sex remains unknown. We hypothesize that males and females with NIDCM respond similarly to MSC therapy. METHODS AND RESULTS Male (n = 24) and female (n = 10) patients from the POSEIDON-DCM trial who received MSCs via transendocardial injections were evaluated over 12 months. Endothelial function was measured at baseline and 3 months post-transendocardial stem cell injection (TESI). At baseline, ejection fraction (EF) was lower (P = 0.004) and end-diastolic volume (EDV; P = 0.0002) and end-systolic volume (ESV; P = 0.0002) were higher in males vs. females. In contrast, baseline demographic characteristics, Minnesota Living with Heart Failure Questionnaire (MLHFQ), and 6-min walk test (6MWT) were similar between groups. EF improved in males by 6.2 units (P = 0.04) and in females by 8.6 units (P = 0.04; males vs. females, P = 0.57). EDV and ESV were unchanged over time. The MLHFQ score, New York Heart Association (NYHA) class, endothelial progenitor cell-colony forming units, and serum tumour necrosis factor alpha improved similarly in both groups. CONCLUSION Despite major differences in phenotypic presentation of NIDCM in males and females, this study is the first of its kind to demonstrate that MSC therapy improves a variety of parameters in NIDCM irrespective of patient sex. These findings have important clinical and pathophysiologic implications regarding the impact of sex on responses to cell-based therapy for NIDCM.
Collapse
Affiliation(s)
- Victoria Florea
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building - 9th Floor 1501 NW 10th Ave, Miami, FL 33136, USA
| | - Angela C Rieger
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building - 9th Floor 1501 NW 10th Ave, Miami, FL 33136, USA
| | - Makoto Natsumeda
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building - 9th Floor 1501 NW 10th Ave, Miami, FL 33136, USA
| | - Bryon A Tompkins
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building - 9th Floor 1501 NW 10th Ave, Miami, FL 33136, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Monisha N Banerjee
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building - 9th Floor 1501 NW 10th Ave, Miami, FL 33136, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Ivonne H Schulman
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building - 9th Floor 1501 NW 10th Ave, Miami, FL 33136, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Courtney Premer
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building - 9th Floor 1501 NW 10th Ave, Miami, FL 33136, USA
| | - Aisha Khan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building - 9th Floor 1501 NW 10th Ave, Miami, FL 33136, USA
| | - Krystalenia Valasaki
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building - 9th Floor 1501 NW 10th Ave, Miami, FL 33136, USA
| | - Bettina Heidecker
- Department of Cardiology, Charite Berlin University of Medicine, Berlin, Germany
| | - Alejandro Mantero
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Wayne Balkan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building - 9th Floor 1501 NW 10th Ave, Miami, FL 33136, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Raul D Mitrani
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building - 9th Floor 1501 NW 10th Ave, Miami, FL 33136, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building - 9th Floor 1501 NW 10th Ave, Miami, FL 33136, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
9
|
Bonsack B, Heyck M, Kingsbury C, Cozene B, Sadanandan N, Lee JY, Borlongan CV. Fast-tracking regenerative medicine for traumatic brain injury. Neural Regen Res 2020; 15:1179-1190. [PMID: 31960797 PMCID: PMC7047809 DOI: 10.4103/1673-5374.270294] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 08/22/2019] [Accepted: 09/26/2019] [Indexed: 12/15/2022] Open
Abstract
Traumatic brain injury remains a global health crisis that spans all demographics, yet there exist limited treatment options that may effectively curtail its lingering symptoms. Traumatic brain injury pathology entails a progression from primary injury to inflammation-mediated secondary cell death. Sequestering this inflammation as a means of ameliorating the greater symptomology of traumatic brain injury has emerged as an attractive treatment prospect. In this review, we recapitulate and evaluate the important developments relating to regulating traumatic brain injury-induced neuroinflammation, edema, and blood-brain barrier disintegration through pharmacotherapy and stem cell transplants. Although these studies of stand-alone treatments have yielded some positive results, more therapeutic outcomes have been documented from the promising area of combined drug and stem cell therapy. Harnessing the facilitatory properties of certain pharmaceuticals with the anti-inflammatory and regenerative effects of stem cell transplants creates a synergistic effect greater than the sum of its parts. The burgeoning evidence in favor of combined drug and stem cell therapies warrants more elaborate preclinical studies on this topic in order to pave the way for later clinical trials.
Collapse
Affiliation(s)
- Brooke Bonsack
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| | - Matt Heyck
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| | - Chase Kingsbury
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| | - Blaise Cozene
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| | - Nadia Sadanandan
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| | - Jea-Young Lee
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| | - Cesar V. Borlongan
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| |
Collapse
|
10
|
Zumwalt M, Reddy AP. Stem Cells for Treatment of Musculoskeletal Conditions - Orthopaedic/Sports Medicine Applications. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165624. [PMID: 31794866 DOI: 10.1016/j.bbadis.2019.165624] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 11/22/2019] [Accepted: 11/23/2019] [Indexed: 12/20/2022]
Abstract
A myriad of musculoskeletal conditions afflicts a vast number of the world's population from birth to death. Countless pathological diseases and traumatic injuries (acute and chronic) contribute to different human disabilities, causing a tremendous financial toll on the economy of healthcare. The medical field is continually searching for novel ways to combat orthopedically related conditions. The immediate goal is the restoration of anatomy then ultimately return of function in hopes of enhancing quality if not the quantity of life. Traditional methods involve surgical correction/reconstruction of skeletal deformities from fractures/soft tissue damage/ruptures or replacement/resection of degenerated joints. Modern research is currently concentrating on innovative procedures to replenish/restore the human body close to its original/natural state [1, 2].
Collapse
Affiliation(s)
- Mimi Zumwalt
- Texas Tech University Health Sciences Center, Department of Orthopaedic Surgery, 3601 4(th) Street STOP 9436, Lubbock, TX 79430 United States of America.
| | - Arubala P Reddy
- Texas Tech University, 1301 Akron Avenue, Lubbock, TX 79409 United States of America.
| |
Collapse
|
11
|
Reddy AP, Ravichandran J, Carkaci-Salli N. Neural regeneration therapies for Alzheimer's and Parkinson's disease-related disorders. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165506. [PMID: 31276770 DOI: 10.1016/j.bbadis.2019.06.020] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 06/24/2019] [Accepted: 06/26/2019] [Indexed: 12/20/2022]
Abstract
Neurodegenerative diseases are devastating mental illnesses without a cure. Alzheimer's disease (AD) characterized by memory loss, multiple cognitive impairments, and changes in personality and behavior. Although tremendous progress has made in understanding the basic biology in disease processes in AD and PD, we still do not have early detectable biomarkers for these diseases. Just in the United States alone, federal and nonfederal funding agencies have spent billions of dollars on clinical trials aimed at finding drugs, but we still do not have a drug or an agent that can slow the AD or PD disease process. One primary reason for this disappointing result may be that the clinical trials enroll patients with AD or PD at advances stages. Although many drugs and agents are tested preclinical and are promising, in human clinical trials, they are mostly ineffective in slowing disease progression. One therapy that has been promising is 'stem cell therapy' based on cell culture and pre-clinical studies. In the few clinical studies that have investigated therapies in clinical trials with AD and PD patients at stage I. The therapies, such as stem cell transplantation - appear to delay the symptoms in AD and PD. The purpose of this article is to describe clinical trials using 1) stem cell transplantation methods in AD and PD mouse models and 2) regenerative medicine in AD and PD mouse models, and 3) the current status of investigating preclinical stem cell transplantation in patients with AD and PD.
Collapse
Affiliation(s)
- Arubala P Reddy
- Pharmacology & Neuroscience Department, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States.
| | - Janani Ravichandran
- Texas Tech University Health Sciences Center El Paso, 5001 El Paso Drive, El Paso, TX 79905, United States.
| | - Nurgul Carkaci-Salli
- Department of Pharmacology, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033.
| |
Collapse
|
12
|
Balzano F, Bellu E, Basoli V, Dei Giudici S, Santaniello S, Cruciani S, Facchin F, Oggiano A, Capobianco G, Dessole F, Ventura C, Dessole S, Maioli M. Lessons from human umbilical cord: gender differences in stem cells from Wharton's jelly. Eur J Obstet Gynecol Reprod Biol 2019; 234:143-148. [PMID: 30690190 DOI: 10.1016/j.ejogrb.2018.12.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 12/12/2018] [Accepted: 12/17/2018] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To study the molecular features of mesenchymal stem cells from Wharton Jelly (WJ-MSCs) of umbilical cord to predict their differentiation capacity. DESIGN Comparison of gene expression from mesenchymal stem cells of male and female umbilical cord SETTING: University hospital PATIENT (S): umbilical cords (n = 12, 6 males and 6 females) retrieved from spontaneous full-term vaginal delivery of healthy women INTERVENTION: we analyzed the expression of the stemness related genes C-MYC, OCT4, SOX2 and NANOG and of the epigenetic modulating gene DNA-methyltransferase 1 (DNMT1). MEAN OUTCOME MEASURE WJ-MSCs were isolated by standard procedures and immunophenotypically characterized. Gene expression analysis of stemness related genes and the epigenetic modulating gene DNMT1 were performed by real-time PCR RESULTS: expression of the OCT4 and DNMT1 genes was significantly higher in WJ- MSCs isolated from male subjects, as compared to MSCs isolated from female-derived WJ. The resulting higher expression of OCT4 and DNMT1 in WJ-MSCs from males as compared with female WJ-MSCs for the first time identifies a specific relationship between stemness genes, an epigenetic modulator, and gender differences. CONCLUSION our findings disclose novel biomedical implications in WJ-MSCs related to the sex of the donor, thus providing additional cues to exploit their regenerative potential in allogenic transplantation.
Collapse
Affiliation(s)
- Francesca Balzano
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
| | - Emanuela Bellu
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
| | - Valentina Basoli
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems - Eldor Lab, Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
| | - Silvia Dei Giudici
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, Sassari 07100, Italy.
| | - Sara Santaniello
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems - Eldor Lab, Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
| | - Sara Cruciani
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems - Eldor Lab, Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
| | - Federica Facchin
- Department of Experimental, Diagnostic and Speciality Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy.
| | - Annalisa Oggiano
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, Sassari 07100, Italy.
| | - Giampiero Capobianco
- Department of Medical, Surgical and experimental Sciences, Gynecologic and Obstetric Clinic, University of Sassari, Italy.
| | - Francesco Dessole
- Department of Medical, Surgical and experimental Sciences, Gynecologic and Obstetric Clinic, University of Sassari, Italy.
| | - Carlo Ventura
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems - Eldor Lab, Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
| | - Salvatore Dessole
- Department of Medical, Surgical and experimental Sciences, Gynecologic and Obstetric Clinic, University of Sassari, Italy.
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems - Eldor Lab, Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy; Center for Developmental Biology and Reprogramming- CEDEBIOR, Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy; Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato, Cagliari, Italy.
| |
Collapse
|
13
|
Fathi-Kazerooni M, Tavoosidana G. Menstrual Blood Stem Cell Transplantation in Mice Model of Acute Liver Failure: Does Gender of Recipient Affect the Outcome? Avicenna J Med Biotechnol 2019; 11:308-316. [PMID: 31908739 PMCID: PMC6925399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
BACKGROUND There exists a dramatic rise in liver failure and numerous patients undergo liver transplant for life-saving reasons annually. Introducing alternatives to allo-graft transplantation is necessary due to present limitations. Recently, a noninvasive stem cell population from Menstrual blood-derived Stem Cells (MenSCs) has been identified. There is an increasing interest in the application of MenSCs in tissue engineering; however, the fact that these gender-specific stem cells are safe for use in male sex is still not well defined. METHODS In this research, a model of acute liver failure was created in male and female immunocompetent Balb-C mice through intraperitoneal injection of Carbon tetrachlo-ride (CCl4 ) and MenSCs were transplanted intravenously 48 hrs after induction of liver injury to evaluate their therapeutic potential. All mice were sacrificed on days 1, 7, and 30 post-transplantation to examine biochemical and molecular markers and pathological appearances. RESULTS Results showed the liver engraftment of MenSCs by immunofluorescence staining using anti-human mitochondrial antibody in both male and female treated groups. The restoration of serum markers of liver injury, aspartate aminotransferase and ala-nine aminotransferase, as well as expression levels of liver-specific genes, tyrosine aminotransferase and cholesterol 7 alpha-hydroxylase, were more significant in the female treated group compared with the male treated group on day 7 (p<0.05); however, after 30 days, there were no significant differences. Furthermore, hematoxylin and eosin and periodic acid-Schiff staining of liver sections demonstrated the considerable liver regeneration post cell therapy in both groups. Notably, data has shown that MenSCs could engraft into injured liver tissues and result in the same effect in the regeneration of liver function in both genders. CONCLUSION Results of this study introduce MenSCs therapy as an attractive alternative approach for liver repairing and regeneration which has no gender constraints.
Collapse
Affiliation(s)
| | - Gholamreza Tavoosidana
- Corresponding author: Gholamreza Tavoosidana, Ph.D. Department of Molecular Medicine School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, P.O. Box 1417755469, Tehran, Iran, Tel: +98 21 88991118, Fax: +98 21 88991117, E-mail:
| |
Collapse
|
14
|
Kaneko T, Gu B, Sone PP, Zaw SYM, Murano H, Zaw ZCT, Okiji T. Dental Pulp Tissue Engineering Using Mesenchymal Stem Cells: a Review with a Protocol. Stem Cell Rev Rep 2018; 14:668-676. [PMID: 29804171 DOI: 10.1007/s12015-018-9826-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mesenchymal stem cells (MSCs) are adult stem cells that can be isolated from human and animal sources such as rats. Recently, an in vivo protocol for pulp tissue engineering using implantation of bone marrow MSCs into rat pulpotomized molars was established by our research group. This coronal pulp regeneration model showed almost complete regeneration/healing with dentin bridge formation when the cavity was sealed with mineral trioxide aggregate (MTA) to create a biocompatible seal of the pulp. This method is a powerful tool for elucidating the processes of dental pulp tissue regeneration following implantation of MSCs. In the present review, we discuss the literature in the field of dental pulp tissue engineering using MSCs including dental pulp stem cells and stem cells from exfoliated deciduous teeth. In addition, we present a brief step-by-step protocol of the coronal pulp regeneration model focusing on the implantation of rat bone marrow MSCs, biodegradable scaffolds, and hydrogels in pulpotomized rat molars. The protocol may lay the foundation for studies aiming at defining further histological and molecular mechanism of the rat pulp tissue engineering.
Collapse
Affiliation(s)
- Tomoatsu Kaneko
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8549, Japan.
| | - Bin Gu
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8549, Japan
| | - Phyo Pyai Sone
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8549, Japan
| | - Su Yee Myo Zaw
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8549, Japan
| | - Hiroki Murano
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8549, Japan
| | - Zar Chi Thein Zaw
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8549, Japan
| | - Takashi Okiji
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8549, Japan
| |
Collapse
|
15
|
Schimmel SJ, Acosta S, Lozano D. Neuroinflammation in traumatic brain injury: A chronic response to an acute injury. Brain Circ 2017; 3:135-142. [PMID: 30276315 PMCID: PMC6057689 DOI: 10.4103/bc.bc_18_17] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 09/02/2017] [Accepted: 09/05/2017] [Indexed: 02/06/2023] Open
Abstract
Every year, approximately 1.4 million US citizens visit emergency rooms for traumatic brain injuries. Formerly known as an acute injury, chronic neurodegenerative symptoms such as compromised motor skills, decreased cognitive abilities, and emotional and behavioral changes have caused the scientific community to consider chronic aspects of the disorder. The injury causing impact prompts multiple cell death processes, starting with neuronal necrosis, and progressing to various secondary cell death mechanisms. Secondary cell death mechanisms, including excitotoxicity, oxidative stress, mitochondrial dysfunction, blood-brain barrier disruption, and inflammation accompany chronic traumatic brain injury (TBI) and often contribute to long-term disabilities. One hallmark of both acute and chronic TBI is neuroinflammation. In acute stages, neuroinflammation is beneficial and stimulates an anti-inflammatory response to the damage. Conversely, in chronic TBI, excessive inflammation stimulates the aforementioned secondary cell death. Converting inflammatory cells from pro-inflammatory to anti-inflammatory may expand the therapeutic window for treating TBI, as inflammation plays a role in all stages of the injury. By expanding current research on the role of inflammation in TBI, treatment options and clinical outcomes for afflicted individuals may improve. This paper is a review article. Referred literature in this paper has been listed in the references section. The data sets supporting the conclusions of this article are available online by searching various databases, including PubMed. Some original points in this article come from the laboratory practice in our research center and the authors' experiences.
Collapse
Affiliation(s)
| | - Sandra Acosta
- Center of Excellence for Aging and Brain, Tampa, FL, USA
| | - Diego Lozano
- School of Medicine, University of Miami School of Medicine, Miami, FL, USA
| |
Collapse
|
16
|
Alvarez CV, Oroz-Gonjar F, Garcia-Lavandeira M. Future perspectives in adult stem cell turnover: Implications for endocrine physiology and disease. Mol Cell Endocrinol 2017; 445:1-6. [PMID: 27956115 DOI: 10.1016/j.mce.2016.12.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 12/08/2016] [Indexed: 02/06/2023]
Affiliation(s)
- Clara V Alvarez
- Centre for Investigations in Molecular Medicine and Chronic Disease (CIMUS) and Institute of Investigaciones Sanitarias (IDIS), Group of Endocrine Neoplasia and Differentiation, University of Santiago de Compostela (USC), Santiago de Compostela, Spain.
| | - Fernando Oroz-Gonjar
- Centre for Investigations in Molecular Medicine and Chronic Disease (CIMUS) and Institute of Investigaciones Sanitarias (IDIS), Group of Endocrine Neoplasia and Differentiation, University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Montserrat Garcia-Lavandeira
- Centre for Investigations in Molecular Medicine and Chronic Disease (CIMUS) and Institute of Investigaciones Sanitarias (IDIS), Group of Endocrine Neoplasia and Differentiation, University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| |
Collapse
|
17
|
Lim S, Kim IK, Choi JW, Seo HH, Lim KH, Lee S, Lee HB, Kim SW, Hwang KC. Gender-dimorphic effects of adipose-derived stromal vascular fractions on HUVECs exposed to oxidative stress. Int J Med Sci 2017; 14:911-919. [PMID: 28824330 PMCID: PMC5562200 DOI: 10.7150/ijms.19998] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 05/17/2017] [Indexed: 12/22/2022] Open
Abstract
Stromal vascular fractions (SVFs) are a heterogeneous collection of cells within adipose tissue that are being studied for various clinical indications. In this study, we aimed to determine whether SVF transplantation into impaired tissues has differential effects on inflammatory and angiogenetic properties with regard to gender. As reactive oxygen species have been implicated in cardiovascular disease development, we investigated differences in gene and protein expression related to inflammation and angiogenesis in HUVECs co-cultured with adipose-derived SVFs from male (M group) and female (F group) individuals under oxidative stress conditions. The expression of several inflammatory (interleukin (IL)-33) and angiogenetic (platelet-derived growth factor (PDGF)) factors differed dramatically between male and female donors. Anti-inflammatory and pro-angiogenetic responses were observed in HUVECs co-cultured with SVFs under oxidative stress conditions, and these characteristics may exhibit partially differential effects according to gender. Using network analysis, we showed that co-culturing HUVECs with SVFs ameliorated pyroptosis/apoptosis via an increase in oxidative stress. Activation of caspase-1 and IL-1B was significantly altered in HUVECs co-cultured with SVFs from female donors. These findings regarding gender-dimorphic regulation of adipose-derived SVFs provide valuable information that can be used for evidence-based gender-specific clinical treatment of SVF transplantation for understanding of cardiovascular disease, allowing for the development of additional treatment.
Collapse
Affiliation(s)
- Soyeon Lim
- Catholic Kwandong University, International St. Mary's Hospital, Incheon Metropolitan City, 22711, Republic of Korea.,Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do 25601, Republic of Korea
| | - Il-Kwon Kim
- Catholic Kwandong University, International St. Mary's Hospital, Incheon Metropolitan City, 22711, Republic of Korea.,Cell Therapy Center, Catholic Kwandong University International St. Mary's Hospital, Incheon Metropolitan City, 22711, Republic of Korea
| | - Jung-Won Choi
- Catholic Kwandong University, International St. Mary's Hospital, Incheon Metropolitan City, 22711, Republic of Korea.,Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do 25601, Republic of Korea
| | - Hyang-Hee Seo
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Kyu Hee Lim
- Department of Veterinary Physiology, College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, Jeonju City, Jeollabuk-Do, Republic of Korea
| | - Seahyoung Lee
- Catholic Kwandong University, International St. Mary's Hospital, Incheon Metropolitan City, 22711, Republic of Korea.,Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do 25601, Republic of Korea
| | - Hoon-Bum Lee
- Catholic Kwandong University, International St. Mary's Hospital, Incheon Metropolitan City, 22711, Republic of Korea.,Department of Plastic and Reconstructive Surgery, Catholic Kwandong University, International St. Mary's Hospital, Incheon Metropolitan City, 22711, Republic of Korea
| | - Sang Woo Kim
- Catholic Kwandong University, International St. Mary's Hospital, Incheon Metropolitan City, 22711, Republic of Korea.,Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do 25601, Republic of Korea
| | - Ki-Chul Hwang
- Catholic Kwandong University, International St. Mary's Hospital, Incheon Metropolitan City, 22711, Republic of Korea.,Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do 25601, Republic of Korea
| |
Collapse
|
18
|
Contreras A, Orozco AF, Resende M, Schutt RC, Traverse JH, Henry TD, Lai D, Cooke JP, Bolli R, Cohen ML, Moyé L, Pepine CJ, Yang PC, Perin EC, Willerson JT, Taylor DA. Identification of cardiovascular risk factors associated with bone marrow cell subsets in patients with STEMI: a biorepository evaluation from the CCTRN TIME and LateTIME clinical trials. Basic Res Cardiol 2017; 112:3. [PMID: 27882430 PMCID: PMC5760218 DOI: 10.1007/s00395-016-0592-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 11/14/2016] [Indexed: 12/13/2022]
Abstract
Autologous bone marrow mononuclear cell (BM-MNC) therapy for patients with ST-segment elevation myocardial infarction (STEMI) has produced inconsistent results, possibly due to BM-MNC product heterogeneity. Patient-specific cardiovascular risk factors (CRFs) may contribute to variations in BM-MNC composition. We sought to identify associations between BM-MNC subset frequencies and specific CRFs in STEMI patients. Bone marrow was collected from 191 STEMI patients enrolled in the CCTRN TIME and LateTIME trials. Relationships between BM-MNC subsets and CRFs were determined with multivariate analyses. An assessment of CRFs showed that hyperlipidemia and hypertension were associated with a higher frequency of CD11b+ cells (P = 0.045 and P = 0.016, respectively). In addition, we found that females had lower frequencies of CD11b+ (P = 0.018) and CD45+CD14+ (P = 0.028) cells than males, age was inversely associated with the frequency of CD45+CD31+ cells (P = 0.001), smoking was associated with a decreased frequency of CD45+CD31+ cells (P = 0.013), glucose level was positively associated with the frequency of CD45+CD3+ cells, and creatinine level (an indicator of renal function) was inversely associated with the frequency of CD45+CD3+ cells (P = 0.015). In conclusion, the frequencies of monocytic, lymphocytic, and angiogenic BM-MNCs varied in relation to patients' CRFs. These phenotypic variations may affect cell therapy outcomes and might be an important consideration when selecting patients for and reviewing results from autologous cell therapy trials.
Collapse
Affiliation(s)
| | | | | | - Robert C Schutt
- Houston Methodist DeBakey Heart and Vascular Center, Houston Methodist Research Institute, Houston, TX, USA
| | - Jay H Traverse
- Minneapolis Heart Institute Foundation, Abbott Northwestern Hospital, Minneapolis, MN, USA
| | | | - Dejian Lai
- UT Health School of Public Health, Houston, TX, USA
| | - John P Cooke
- Houston Methodist DeBakey Heart and Vascular Center, Houston Methodist Research Institute, Houston, TX, USA
| | | | | | - Lem Moyé
- UT Health School of Public Health, Houston, TX, USA.
| | - Carl J Pepine
- College of Medicine, University of Florida, Gainesville, FL, USA
| | - Phillip C Yang
- Stanford University School of Medicine, Stanford, CA, USA
| | | | | | | |
Collapse
|
19
|
Distinct and Shared Determinants of Cardiomyocyte Contractility in Multi-Lineage Competent Ethnically Diverse Human iPSCs. Sci Rep 2016; 6:37637. [PMID: 27917881 PMCID: PMC5137163 DOI: 10.1038/srep37637] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 10/31/2016] [Indexed: 12/28/2022] Open
Abstract
The realization of personalized medicine through human induced pluripotent stem cell (iPSC) technology can be advanced by transcriptomics, epigenomics, and bioinformatics that inform on genetic pathways directing tissue development and function. When possible, population diversity should be included in new studies as resources become available. Previously we derived replicate iPSC lines of African American, Hispanic-Latino and Asian self-designated ethnically diverse (ED) origins with normal karyotype, verified teratoma formation, pluripotency biomarkers, and tri-lineage in vitro commitment. Here we perform bioinformatics of RNA-Seq and ChIP-seq pluripotency data sets for two replicate Asian and Hispanic-Latino ED-iPSC lines that reveal differences in generation of contractile cardiomyocytes but similar and robust differentiation to multiple neural, pancreatic, and smooth muscle cell types. We identify shared and distinct genes and contributing pathways in the replicate ED-iPSC lines to enhance our ability to understand how reprogramming to iPSC impacts genes and pathways contributing to cardiomyocyte contractility potential.
Collapse
|
20
|
Nagamura F. The Importance of Recruiting a Diverse Population for Stem Cell Clinical Trials. CURRENT STEM CELL REPORTS 2016. [DOI: 10.1007/s40778-016-0062-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
21
|
Mashkouri S, Crowley MG, Liska MG, Corey S, Borlongan CV. Utilizing pharmacotherapy and mesenchymal stem cell therapy to reduce inflammation following traumatic brain injury. Neural Regen Res 2016; 11:1379-1384. [PMID: 27857726 PMCID: PMC5090825 DOI: 10.4103/1673-5374.191197] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
The pathologic process of chronic phase traumatic brain injury is associated with spreading inflammation, cell death, and neural dysfunction. It is thought that sequestration of inflammatory mediators can facilitate recovery and promote an environment that fosters cellular regeneration. Studies have targeted post-traumatic brain injury inflammation with the use of pharmacotherapy and cell therapy. These therapeutic options are aimed at reducing the edematous and neurodegenerative inflammation that have been associated with compromising the integrity of the blood-brain barrier. Although studies have yielded positive results from anti-inflammatory pharmacotherapy and cell therapy individually, emerging research has begun to target inflammation using combination therapy. The joint use of anti-inflammatory drugs alongside stem cell transplantation may provide better clinical outcomes for traumatic brain injury patients. Despite the promising results in this field of research, it is important to note that most of the studies mentioned in this review have completed their studies using animal models. Translation of this research into a clinical setting will require additional laboratory experiments and larger preclinical trials.
Collapse
Affiliation(s)
- Sherwin Mashkouri
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Marci G Crowley
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Michael G Liska
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Sydney Corey
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| |
Collapse
|
22
|
Schimke MM, Marozin S, Lepperdinger G. Patient-Specific Age: The Other Side of the Coin in Advanced Mesenchymal Stem Cell Therapy. Front Physiol 2015; 6:362. [PMID: 26696897 PMCID: PMC4667069 DOI: 10.3389/fphys.2015.00362] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 11/16/2015] [Indexed: 12/12/2022] Open
Abstract
Multipotential mesenchymal stromal cells (MSC) are present as a rare subpopulation within any type of stroma in the body of higher animals. Prominently, MSC have been recognized to reside in perivascular locations, supposedly maintaining blood vessel integrity. During tissue damage and injury, MSC/pericytes become activated, evade from their perivascular niche and are thus assumed to support wound healing and tissue regeneration. In vitro MSC exhibit demonstrated capabilities to differentiate into a wide variety of tissue cell types. Hence, many MSC-based therapeutic approaches have been performed to address bone, cartilage, or heart regeneration. Furthermore, prominent studies showed efficacy of ex vivo expanded MSC to countervail graft-vs.-host-disease. Therefore, additional fields of application are presently conceived, in which MSC-based therapies potentially unfold beneficial effects, such as amelioration of non-healing conditions after tendon or spinal cord injury, as well as neuropathies. Working along these lines, MSC-based scientific research has been forged ahead to prominently occupy the clinical stage. Aging is to a great deal stochastic by nature bringing forth changes in an individual fashion. Yet, is aging of stem cells or/and their corresponding niche considered a determining factor for outcome and success of clinical therapies?
Collapse
Affiliation(s)
- Magdalena M Schimke
- Department of Cell Biology and Physiology, Stem Cell Research, Aging and Regeneration, University Salzburg Salzburg, Austria
| | - Sabrina Marozin
- Department of Cell Biology and Physiology, Stem Cell Research, Aging and Regeneration, University Salzburg Salzburg, Austria
| | - Günter Lepperdinger
- Department of Cell Biology and Physiology, Stem Cell Research, Aging and Regeneration, University Salzburg Salzburg, Austria
| |
Collapse
|
23
|
Lozano D, Gonzales-Portillo GS, Acosta S, de la Pena I, Tajiri N, Kaneko Y, Borlongan CV. Neuroinflammatory responses to traumatic brain injury: etiology, clinical consequences, and therapeutic opportunities. Neuropsychiatr Dis Treat 2015; 11:97-106. [PMID: 25657582 PMCID: PMC4295534 DOI: 10.2147/ndt.s65815] [Citation(s) in RCA: 228] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Traumatic brain injury (TBI) is a serious public health problem accounting for 1.4 million emergency room visits by US citizens each year. Although TBI has been traditionally considered an acute injury, chronic symptoms reminiscent of neurodegenerative disorders have now been recognized. These progressive neurodegenerative-like symptoms manifest as impaired motor and cognitive skills, as well as stress, anxiety, and mood affective behavioral alterations. TBI, characterized by external bumps or blows to the head exceeding the brain's protective capacity, causes physical damage to the central nervous system with accompanying neurological dysfunctions. The primary impact results in direct neural cell loss predominantly exhibiting necrotic death, which is then followed by a wave of secondary injury cascades including excitotoxicity, oxidative stress, mitochondrial dysfunction, blood-brain barrier disruption, and inflammation. All these processes exacerbate the damage, worsen the clinical outcomes, and persist as an evolving pathological hallmark of what we now describe as chronic TBI. Neuroinflammation in the acute stage of TBI mobilizes immune cells, astrocytes, cytokines, and chemokines toward the site of injury to mount an antiinflammatory response against brain damage; however, in the chronic stage, excess activation of these inflammatory elements contributes to an "inflamed" brain microenvironment that principally contributes to secondary cell death in TBI. Modulating these inflammatory cells by changing their phenotype from proinflammatory to antiinflammatory would likely promote therapeutic effects on TBI. Because neuroinflammation occurs at acute and chronic stages after the primary insult in TBI, a treatment targeting neuroinflammation may have a wider therapeutic window for TBI. To this end, a better understanding of TBI etiology and clinical manifestations, especially the pathological presentation of chronic TBI with neuroinflammation as a major component, will advance our knowledge on inflammation-based disease mechanisms and treatments.
Collapse
Affiliation(s)
- Diego Lozano
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Gabriel S Gonzales-Portillo
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Sandra Acosta
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Ike de la Pena
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Naoki Tajiri
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Yuji Kaneko
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| |
Collapse
|