Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Andrew T. Crane is active.

Publication


Featured researches published by Andrew T. Crane.


Stem Cells | 2014

Transplants of Adult Mesenchymal and Neural Stem Cells Provide Neuroprotection and Behavioral Sparing in a Transgenic Rat Model of Huntington's Disease

Julien Rossignol; Kyle D. Fink; Kendra K Davis; Steven Clerc; Andrew T. Crane; Jessica Matchynski; Steven A. Lowrance; Matthew C. Bombard; Nicholas W. Dekorver; Laurent Lescaudron; Gary L. Dunbar

Stem cells have gained significant interest as a potential treatment of neurodegenerative diseases, including Huntingtons disease (HD). One source of these cells is adult neural stem cells (aNSCs), which differentiate easily into neuronal lineages. However, these cells are vulnerable to immune responses following transplantation. Another source is bone‐marrow‐derived mesenchymal stem cells (MSCs), which release neurotrophic factors and anti‐inflammatory cytokines following transplantation, and are less vulnerable to rejection. The goal of this study was to compare the efficacy of transplants of MSCs, aNSCs, or cotransplants of MSCs and aNSCs for reducing deficits in a transgenic rat model of HD. HD rats received intrastriatal transplantations of 400,000 MSCs, aNSCs, or a combination of MSCs/aNSCs, while wild‐type and HD controls were given vehicle. Rats were tested on the rotarod over the course of 20 weeks. The results indicated that transplants of: (a) aNSCs produced a strong immune response and conferred short‐term behavioral benefits; (b) MSCs elicited a relatively weak immune response, and provided a longer term behavioral benefit; and (c) combined MSCs and aNSCs conferred long‐term behavioral benefits and increased survival of the transplanted aNSCs. The finding that cotransplanting MSCs with aNSCs can prolong aNSC survival and provide greater behavioral sparing than when the transplants contains only aNSCs suggests that MSCs are capable of creating a more suitable microenvironment for aNSC survival. This cotransplantation strategy may be useful as a future therapeutic option for treating HD, especially if long‐term survival of differentiated cells proves to be critically important for preserving lasting functional outcomes. Stem Cells 2014;32:500–509


Restorative Neurology and Neuroscience | 2012

The effects of acute voluntary wheel running on recovery of function following medial frontal cortical contusions in rats.

Andrew T. Crane; Kyle D. Fink; Jeffrey S. Smith

PURPOSE Traumatic brain injury (TBI) produces significant deficits in executive function, sensory-motor function, and on spatial learning tasks. We wish to study if recovery from TBI can be benefited by voluntary exercise. METHODS A variation of the stop-signal reaction time (SSRT) task was employed to measure rats ability to obtain maximum reinforcers in a complex behavioral task. A 2 × 2 (lesion × treatment) experimental design was constructed with 31 weight restricted male Long-Evans rats which received either bilateral cortical contusions to the medial frontal cortex or sham preparations following the acquisition of the SSRT task (matched based on pre-surgical performance). Following surgery, rats were randomly assigned to either an environment with free access to running wheels or traditional single housing without running wheels. RESULTS Rats receiving a bilateral TBI performed significantly worse than sham operated rats on a complex task. Contrary to our original hypothesis, acute exercise following injury exacerbated the deficits in the complex task that did not return to levels of the injured rats without access to running wheels until post-TBI day 13. CONCLUSION We found a significant interaction between severe bilateral TBI and the introduction of voluntary exercise immediately post-injury. In this paradigm, voluntary wheel running exacerbated the TBI-induced deficit, rather than reducing it.


Stem Cells Translational Medicine | 2014

Intrastriatal Transplantation of Adenovirus-Generated Induced Pluripotent Stem Cells for Treating Neuropathological and Functional Deficits in a Rodent Model of Huntington’s Disease

Kyle D. Fink; Andrew T. Crane; Xavier Lévêque; Dylan J. Dues; Lucas D. Huffman; Allison C. Moore; Darren Story; Rachel E. DeJonge; Aaron Antcliff; Phillip A. Starski; Ming Lu; Laurent Lescaudron; Julien Rossignol; Gary L. Dunbar

Induced pluripotent stem cells (iPSCs) show considerable promise for cell replacement therapies for Huntingtons disease (HD). Our laboratory has demonstrated that tail‐tip fibroblasts, reprogrammed into iPSCs via two adenoviruses, can survive and differentiate into neuronal lineages following transplantation into healthy adult rats. However, the ability of these cells to survive, differentiate, and restore function in a damaged brain is unknown. To this end, adult rats received a regimen of 3‐nitropropionic acid (3‐NP) to induce behavioral and neuropathological deficits that resemble HD. At 7, 21, and 42 days after the initiation of 3‐NP or vehicle, the rats received intrastriatal bilateral transplantation of iPSCs. All rats that received 3‐NP and vehicle treatment displayed significant motor impairment, whereas those that received iPSC transplantation after 3‐NP treatment had preserved motor function. Histological analysis of the brains of these rats revealed significant decreases in optical densitometric measures in the striatum, lateral ventricle enlargement, as well as an increase in striosome size in all rats receiving 3‐NP when compared with sham rats. The 3‐NP‐treated rats given transplants of iPSCs in the 7‐ or 21‐day groups did not exhibit these deficits. Transplantation of iPSCs at the late‐stage (42‐day) time point did not protect against the 3‐NP‐induced neuropathology, despite preserving motor function. Transplanted iPSCs were found to survive and differentiate into region‐specific neurons in the striatum of 3‐NP rats, at all transplantation time points. Taken together, these results suggest that transplantation of adenovirus‐generated iPSCs may provide a potential avenue for therapeutic treatment of HD.


Cell Transplantation | 2014

Survival and differentiation of adenovirus-generated induced pluripotent stem cells transplanted into the rat striatum.

Kyle D. Fink; Julien Rossignol; Ming Lu; Xavier Lévêque; Travis D. Hulse; Andrew T. Crane; Véronique Nerrière-Daguin; Robert D. Wyse; Phillip A. Starski; Matthew T. Schloop; Dylan J. Dues; Steve J. Witte; Cheng Song; Ludovic Vallier; Tuan H. Nguyen; Philippe Naveilhan; Ignacio Anegon; Laurent Lescaudron; Gary L. Dunbar

Induced pluripotent stem cells (iPSCs) offer certain advantages over embryonic stem cells in cell replacement therapy for a variety of neurological disorders. However, reliable procedures, whereby transplanted iPSCs can survive and differentiate into functional neurons, without forming tumors, have yet to be devised. Currently, retroviral or lentiviral reprogramming methods are often used to reprogram somatic cells. Although the use of these viruses has proven to be effective, formation of tumors often results following in vivo transplantation, possibly due to the integration of the reprogramming genes. The goal of the current study was to develop a new approach, using an adenovirus for reprogramming cells, characterize the iPSCs in vitro, and test their safety, survivability, and ability to differentiate into region-appropriate neurons following transplantation into the rat brain. To this end, iPSCs were derived from bone marrow-derived mesenchymal stem cells and tail-tip fibroblasts using a single cassette lentivirus or a combination of adenoviruses. The reprogramming efficiency and levels of pluripotency were compared using immunocytochemistry, flow cytometry, and real-time polymerase chain reaction. Our data indicate that adenovirus-generated iPSCs from tail-tip fibroblasts are as efficient as the method we used for lentiviral reprogramming. All generated iPSCs were also capable of differentiating into neuronal-like cells in vitro. To test the in vivo survivability and the ability to differentiate into region-specific neurons in the absence of tumor formation, 400,000 of the iPSCs derived from tail-tip fibroblasts that were transfected with the adenovirus pair were transplanted into the striatum of adult, immune-competent rats. We observed that these iPSCs produced region-specific neuronal phenotypes, in the absence of tumor formation, at 90 days posttransplantation. These results suggest that adenovirus-generated iPSCs may provide a safe and viable means for neuronal replacement therapies.


Behavioral Neuroscience | 2012

Early cognitive dysfunction in the HD 51 CAG transgenic rat model of Huntington's disease.

Kyle D. Fink; Julien Rossignol; Andrew T. Crane; Kendra K Davis; Angela M Bavar; Nicholas W. Dekorver; Steven A. Lowrance; Mark P. Reilly; Michael I. Sandstrom; Stephan von Hörsten; Laurent Lescaudron; Gary L. Dunbar

Huntingtons disease (HD) is a neurodegenerative disorder in humans caused by an expansion of a CAG trinucleotide repeat that produces choreic movements, which are preceded by cognitive deficits. The HD transgenic rat (tgHD), which contains the human HD mutation with a 51 CAG repeat allele, exhibits motor deficits that begin when these rats are 12 months of age. However, there are no reports of cognitive dysfunction occurring prior to this. To assess whether cognitive dysfunction might precede motor deficits in tgHD rats, one group of 9-month-old male rats with homozygotic mutated genes and one group of wild-type (WT) rats underwent three testing phases in a unique Spatial Operant Reversal Test (SORT) paradigm, as well as assessment of spontaneous motor activity. After testing, morphological and histological examination of the brains were made. Results indicated that tgHD rats acquired the cued-response (Phase 1) portion of the SORT, but made significantly more errors during the reversal (Phase 2) and during the pseudorandomized reversals (Phase 3) portion of the study, when compared to WT rats. Analysis of the data using mathematical principles of reinforcement revealed no memory, motor, or motivational deficits. These results indicate that early cognitive dysfunction, as measured by the SORT, occur prior to motor deficits, gross anatomical changes, or cell loss in the tgHD rat with 51 CAG repeats, and suggest that this protocol could provide a useful screen for therapeutic studies.


Brain Sciences | 2014

Use of Genetically Altered Stem Cells for the Treatment of Huntington’s Disease

Andrew T. Crane; Julien Rossignol; Gary L. Dunbar

Transplantation of stem cells for the treatment of Huntington’s disease (HD) garnered much attention prior to the turn of the century. Several studies using mesenchymal stem cells (MSCs) have indicated that these cells have enormous therapeutic potential in HD and other disorders. Advantages of using MSCs for cell therapies include their ease of isolation, rapid propagation in culture, and favorable immunomodulatory profiles. However, the lack of consistent neuronal differentiation of transplanted MSCs has limited their therapeutic efficacy to slowing the progression of HD-like symptoms in animal models of HD. The use of MSCs which have been genetically altered to overexpress brain derived neurotrophic factor to enhance support of surviving cells in a rodent model of HD provides proof-of-principle that these cells may provide such prophylactic benefits. New techniques that may prove useful for cell replacement therapies in HD include the use of genetically altering fate-restricted cells to produce induced pluripotent stem cells (iPSCs). These iPSCs appear to have certain advantages over the use of embryonic stem cells, including being readily available, easy to obtain, less evidence of tumor formation, and a reduced immune response following their transplantation. Recently, transplants of iPSCs have shown to differentiate into region-specific neurons in an animal model of HD. The overall successes of using genetically altered stem cells for reducing neuropathological and behavioral deficits in rodent models of HD suggest that these approaches have considerable potential for clinical use. However, the choice of what type of genetically altered stem cell to use for transplantation is dependent on the stage of HD and whether the end-goal is preserving endogenous neurons in early-stage HD, or replacing the lost neurons in late-stage HD. This review will discuss the current state of stem cell technology for treating the different stages of HD and possible future directions for stem-cell therapy in HD.


Restorative Neurology and Neuroscience | 2015

Bone-marrow-derived mesenchymal stem cells attenuate cognitive deficits in an endothelin-1 rat model of stroke.

S.A. Lowrance; Kyle D. Fink; Andrew T. Crane; J. Matyas; N.D. Dey; J.J. Matchynski; T. Thibo; T. Reinke; J. Kippe; C. Hoffman; Michael I. Sandstrom; Julien Rossignol; Gary L. Dunbar

PURPOSE Stroke is the third leading cause of death and permanent disability in the United States, often producing long-term cognitive impairments, which are not easily recapitulated in animal models. The goals of this study were to assess whether: (1) the endothelin-1 (ET-1) model of chronic stroke produced discernable cognitive deficits; (2) a spatial operant reversal task (SORT) would accurately measure memory deficits in this model; and (3) bone-marrow-derived mesenchymal stem cells (BMMSCs) could reduce any observed deficits. METHODS Rats were given unilateral intracerebral injections of vehicle or ET-1, a stroke-inducing agent, near the middle cerebral artery. Seven days later, they were given intrastriatal injections of BMMSCs or vehicle, near the ischemic penumbra. The cognitive abilities of the rats were assessed on a novel SORT, which was designed to efficiently distinguish cognitive deficits from potential motoric confounds. RESULTS Rats given ET-1 had significantly more cognitive errors at six weeks post-stroke on the SORT, and that these deficits were attenuated by BMMSC transplants. CONCLUSIONS These findings indicate that: (1) the ET-1 model produces chronic cognitive deficits; (2) the SORT efficiently measures cognitive deficits that are not confounded by motoric impairment; and (3) BMMSCs may be a viable treatment for stroke-induced cognitive dysfunction.


Cell Transplantation | 2016

Mesenchymal Stem Cells as Treatment for Behavioral Deficits and Neuropathology in the 5xFAD Mouse Model of Alzheimer's Disease.

Jessica J. Matchynski-Franks; Colleen Pappas; Julien Rossignol; Tiffany Reinke; Kyle D. Fink; Andrew T. Crane; Alison Twite; Steven A. Lowrance; Cheng Song; Gary L. Dunbar

Alzheimers disease (AD) is characterized by a progressive loss of memory and other cognitive disturbances. The neuropathology of AD includes the major hallmarks of toxic amyloid-β oligomer accumulation and neurofibrillary tangles, as well as increased oxidative stress, cholinergic dysfunction, synapse loss, changes in endogenous neurotrophic factors, and overall degeneration of the brain. Adult mesenchymal stem cells (MSCs) offer the potential for a readily available treatment that would be long lasting, have low likelihood of rejection, and could target a variety of pathological deficits. MSCs have been shown to be effective in alleviating symptoms in some transgenic models of AD, but the optimal location for transplanting MSCs has yet to be determined. In the present study, the behavioral effects of transplantation of MSCs into the lateral ventricles, the hippocampus, or both of these regions were compared in the 5xFAD mouse model of AD. The results indicate that MSC transplants effectively reduce learning deficits in the 5xFAD mouse model and demonstrate a clear impact of MSCs on the levels of Aβ42 in the brains of 5xFAD mice. Overall, these findings support the hypothesis that MSCs may be a viable treatment for AD, especially when injected into the lateral ventricles.


Journal of Stem Cell Research & Therapy | 2014

Will Undifferentiated Induced Pluripotent Stem Cells Ever have Clinical Utility

Julien Rossignol; Andrew T. Crane; Kyle D. Fink; Gary L. Dunbar

The emergence of induced pluripotent stem cell (iPSC) technology, with the capability of iPSCs to differentiate into any type of cell, has advanced the field of stem cell therapies. As the field has progressed towards pre-clinical transplantation of iPSCs, polarizing views of the tumorigenic potential of undifferentiated iPSCs has left many researchers believing that there is no future in the clinical utility of transplanting undifferentiated iPSCs. The potential for insertional mutagenesis and the integration of oncogenes in iPSCs, as well as the teratoma assay in nude mice, has fueled the rationale for one side of the argument, while some iPSC transplantation studies into healthy, immunocompetent and animals have provided evidence that clinical utility is possible. This brief review highlights the perspectives of both sides of the debate while providing representative examples of iPSCs studies, as well as possible safeguards against iPSC-induced tumor formation.


Frontiers in Neuroscience | 2017

Induced Pluripotent Stem Cell-Derived Neural Stem Cell Transplantations Reduced Behavioral Deficits and Ameliorated Neuropathological Changes in YAC128 Mouse Model of Huntington's Disease

Abeer Al-Gharaibeh; Rebecca Culver; Andrew N. Stewart; Bhairavi Srinageshwar; Kristin Spelde; Laura Frollo; Nivya Kolli; Darren Story; Leela Paladugu; Sarah Anwar; Andrew T. Crane; Robert D. Wyse; Panchanan Maiti; Gary L. Dunbar; Julien Rossignol

Huntingtons disease (HD) is a genetic neurodegenerative disorder characterized by neuronal loss and motor dysfunction. Although there is no effective treatment, stem cell transplantation offers a promising therapeutic strategy, but the safety and efficacy of this approach needs to be optimized. The purpose of this study was to test the potential of intra-striatal transplantation of induced pluripotent stem cell-derived neural stem cells (iPS-NSCs) for treating HD. For this purpose, we developed mouse adenovirus-generated iPSCs, differentiated them into neural stem cells in vitro, labeled them with Hoechst, and transplanted them bilaterally into striata of 10-month old wild type (WT) and HD YAC128 mice. We assessed the efficiency of these transplanted iPS-NSCs to reduce motor deficits in YAC128 mice by testing them on an accelerating rotarod task at 1 day prior to transplantation, and then weekly for 10 weeks. Our results showed an amelioration of locomotor deficits in YAC128 mice that received iPS-NSC transplantations. Following testing, the mice were sacrificed, and their brains were analyzed using immunohistochemistry and Western blot (WB). The results from our histological examinations revealed no signs of tumors and evidence that many iPS-NSCs survived and differentiated into region-specific neurons (medium spiny neurons) in both WT and HD mice, as confirmed by co-labeling of Hoechst-labeled transplanted cells with NeuN and DARPP-32. Also, counts of Hoechst-labeled cells revealed that a higher proportion were co-labeled with DARPP-32 and NeuN in HD-, compared to WT- mice, suggesting a dissimilar differentiation pattern in HD mice. Whereas significant decreases were found in counts of NeuN- and DARPP-32-labeled cells, and for neuronal density measures in striata of HD vehicle controls, such decrements were not observed in the iPS-NSCs-transplanted-HD mice. WB analysis showed increase of BDNF and TrkB levels in striata of transplanted HD mice compared to HD vehicle controls. Collectively, our data suggest that iPS-NSCs may provide an effective option for neuronal replacement therapy in HD.

Collaboration


Dive into the Andrew T. Crane's collaboration.

Top Co-Authors

Avatar

Gary L. Dunbar

Central Michigan University

View shared research outputs
Top Co-Authors

Avatar

Julien Rossignol

Central Michigan University

View shared research outputs
Top Co-Authors

Avatar

Kyle D. Fink

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Steven A. Lowrance

Central Michigan University

View shared research outputs
Top Co-Authors

Avatar

Cheng Song

Central Michigan University

View shared research outputs
Top Co-Authors

Avatar

Kendra K Davis

Central Michigan University

View shared research outputs
Top Co-Authors

Avatar

Angela M Bavar

Central Michigan University

View shared research outputs
Top Co-Authors

Avatar

Matthew C. Bombard

Central Michigan University

View shared research outputs
Top Co-Authors

Avatar

Steven Clerc

Central Michigan University

View shared research outputs
Researchain Logo
Decentralizing Knowledge