目前分類:Cell 細胞 (179)

瀏覽方式: 標題列表 簡短摘要

Rodolfa01  

Inducing pluripotency1

Kit T Rodolfa,

Harvard Stem Cell Institute, Stowers Medical Institute, Department of Stem Cell and Regenerative Biology, Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA

http://www.stembook.org/node/514

 

The prospect of personalized regenerative medicine promises to provide treatments for a wide range of degenerative diseases and medical conditions. An important first step in attaining this goal is the production of Pluripotent stem cells directly from individual patients, thereby providing autologous material which, after correcting intrinsic genetic defects and differentiation into required cell types or tissues, could be transplanted into the patient. This chapter reviews the current progress towards this first step, focusing on the techniques used to generate pluripotent cells, the advantages that each offers and the challenges that must be overcome.

 

1. Introduction

Much of the hope invested in embryonic stemEScell research surrounds its promise to provide a broad spectrum of medical applications. The development of such treatments relies on the production of pluripotent stem cells genetically identical to patients. Such stem cells, following differentiation into the disease-relevant cell types, would serve as the key substrate for disease models to study the patient's condition, drug discovery to slow or stop cellular degeneration, and cell replacement therapies after any intrinsic disease-causing genetic defects were repairedsee Figure 1. Beyond providing a renewable source of material that might be transplanted into a patient without suppressing their immune system, the generation of Autologous pluripotent stem cells provides two key advantages over the large number of existing stem cell lines. First, evidence exists that off-target effects of immunosuppressant drugs commonly used in tissue and organ transplants may directly interfere with the function of transplanted tissues, such as the inhibition of β-cell replication that has been observed with drugs used in the treatment of Type I DiabetesNir et al., 2007.

 

Figure 1. The steps of regenerative medicine.

 

The production of cellular therapies requires the optimization of four steps

First, Isolating and Culturing cells that can be readily obtained from a patient in a non-invasive fashion.

Second, the Reprogramming of these cells into a Pluripotent state.

Third, the Directed differentiation of those patient-specific pluripotent cells into the Cell type relevant to their disease.

And, fourth, Techniques for Repairing any intrinsic disease-causing genetic defects and Transplantation of the repaired, differentiated cells into the patient.

Notably, these disease-relevant patient cells can also be used for in vitro disease modeling which may yield new insights into disease mechanisms and drug discovery.

Charlie 發表在 痞客邦 留言(0) 人氣()

小五折圖20100531194320_853_1image003  

幹細胞(Stem cell

名詞解釋-作者:杜蕙蓉 | 中時電子報 – 2014226

工商時報【杜蕙蓉】

 

幹細胞(Stem cell)最簡單的定義是指未充分分化的細胞,具有再生各種組織器官和修復、抗老等潛在功能。

對哺乳動物而言,幹細胞分為兩大類:胚胎幹細胞與成體幹細胞,胚胎幹細胞取自囊胚裡的內細胞團;而成體幹細胞則來自各式各樣的組織,例如:骨髓幹細胞、造血幹細胞、神經幹細胞等。

就產業應用面來看,目前有胚胎幹細胞和臍帶血最被熟知,但胚胎幹細胞不能流通。宣昶有表示,胚胎分裂成16個細胞後,行成胚囊,叫做全能幹細胞,可以變成身體所有的器官、組織,然後才慢慢分化成中胚層、內胚層及外胚層細胞。其中,臍帶血的幹細胞來自內胚層,必須要配對,主要用於治療血液疾病;宣捷使用的幹細胞是中胚層的間質幹細胞,主要來源是胎盤,不僅幹細胞數量多,且不需要配對,在應用廣泛中,對國內再生醫學的發展將有正面助益。

Charlie 發表在 痞客邦 留言(1) 人氣()

1479-5876-7-29-2  

Non-expanded adipose Stromal vascular fraction cell therapy for Multiple sclerosis2

#233

Second patient: A 32-year-old man was diagnosed in 2001 with relapsing-remitting MS, presenting with fatigue and depression, uneven walk pattern, cognitive dysfunction, and a progressive decline in his memory without any specific neurological symptoms. In 2002 he was started on weekly intramuscular Avonex (IFN-b1a, Biogen Idec) and has had no further exacerbations and no evidence of progressive deterioration. Patient's fatigue was treated well with Provigil, and his mood improved significantly due to treatment with Wellbutrin SR. In 2007, the patient complained of some mood changes, with more agitation, irritability, mood destabilization, and cognitive slowing. As depression was suspected in playing a central role in patient's condition, Razadyne was added to the antidepressant regimen.

In 2008, the patient was treated with two I.V. infusions of 25 million autologous adipose-derived SVF cells and multiple intrathecal and intravenous infusions of allogeneic CD34+ and MSC cells. MSC were third party unmatched and CD34 were matched by mixed lymphocyte reaction. All infusions were performed within a 10-day period and were very well tolerated without any significant side effects. The treatment plan also included physical therapy sessions.

Three months after the stem cell infusions the patient reported a significant improvement of his balance and coordination as well as an improved energy level and mood. New MRI images, obtained 7 months after the stem cell treatment showed lesions, very similar to the lesions observed before the stem cell treatment (Figure 2). Currently, he is not taking any antidepressants and is reporting a significantly improved overall condition. His current treatment regiment includes a weekly injection of Avonex, vitamins, minerals and Omega 3.

 

Figure 2. MRI Images obtained before (Panels A and B), and seven months after (Panel C) the stem cell treatment of patient 2. Panels A and B: Consecutive axial FLuid-Attenuated Inversion Recovery (FLAIR) images through the lateral ventricles show multiple small patches of bright signal in the periventricular and subcortical white matter, consistent with plaques of multiple sclerosis. Panel C: Axial FLAIR image shows no significant change in the multiple periventricular and subcortical white-matter plaques. (For the comparison, note that this slice is positioned similar to slice A but at slightly different scanning-angle, so it includes lesions of both slices A and B.).

 

#255

The patient was diagnosed with relapsing-remitting MS in 1993, presenting symptoms were noticeable tingling and burning sensation in the right leg, followed by paraplegia lasting almost three weeks. Neurological investigations at the time uncovered MRI findings suggestive for a demyelinating syndrome. In June of 2008, the patient was treated with two I.V. infusions of 75 million autologous adipose-derived SVF cells and multiple intrathecal and intravenous infusions of allogeneic CD34+ and MSC cells. MSC were third party unmatched and CD34 were matched by mixed lymphocyte reaction. All infusions were performed within a 10-day period and were very well tolerated without any significant side effects. His gait, balance and coordination improved dramatically oven a period of several weeks. His condition continued to improve over the next few months and he is currently reporting a still continuing improvement and ability to jog, run and bike for extended periods of time daily.

 

Conclusion

The patients treated were part of a compassionate-use evaluation of stem cell therapeutic protocols in a physician-initiated manner. Previous experiences in MS patients using allogeneic CD34+ cord blood cells together with MSC did not routinely result in substantial improvements observed in the three cases described above. While obviously no conclusions in terms of therapeutic efficacy can be drawn from the above reports, we believe that further clinical evaluation of autologous SVF cells is warranted in autoimmune conditions.

 

Competing interests

Thomas E Ichim and Neil H Riordan are management and shareholders of Medistem Inc, a company that has filed intellectual property on the use of adipose stromal vascular fraction cells for immune modulation.

 

Authors' contributions

All authors read and approved the final manuscript. NHR, TEI, WPM, HW, FS, FL, MA, JPR, RJH, ANP, MPM, RRL and BM conceived experiments, interpreted data, and wrote the manuscript.

 

Acknowledgements

We thank Victoria Dardov, Rosalia De Necochea Campion, Florica Batu, and Boris Markosian for stimulating discussions.

Charlie 發表在 痞客邦 留言(0) 人氣()

1479-5876-7-29-1  

Non-expanded adipose Stromal vascular fraction cell therapy for Multiple sclerosis1

Neil H Riordan1, Thomas E Ichim1*, Wei-Ping Min2, Hao Wang2, Fabio Solano3, Fabian Lara3, Miguel Alfaro4, Jorge Paz Rodriguez5, Robert J Harman6, Amit N Patel7, Michael P Murphy8, Roland R Lee109 and Boris Minev1112

*Corresponding author: Thomas E Ichim thomas.ichim@gmail.com

Author Affiliations

1 Medistem Inc, San Diego, CA, USA

2 Department of Surgery, University of Western Ontario, London, Ontario, Canada

3 Cell Medicine Institutes, San Jose, Costa Rica

4 Hospital CIMA, San Jose, Costa Rica

5 Cell Medicine Institutes, Panama City, Panama

6 Vet-Stem, Inc. Poway, CA, USA

7 Dept of Cardiothoracic Surgery, University of Utah, Salt Lake City, Utah, USA

8 Division of Medicine, Indiana University School of Medicine, Indiana, USA

9 Department of Radiology, University of Canlfornia San Diego, San Diego, CA, USA

10 Veterans Administration, San Diego, CA, USA

11 Moores Cancer Center, University of California, San Diego, CA, USA

12 Department of Medicine, Division of Neurosurgery, University of California San Diego, San Diego, CA, USA

 

Journal of Translational Medicine 2009, 7:29

doi:10.1186/1479-5876-7-29

 

 

The electronic version of this article is the complete one and can be found online at : http://www.translational-medicine.com/content/7/1/29.

Received : 16 March 2009

Accepted : 24 April 2009

Published : 24 April 2009

© 2009 Riordan et al; licensee BioMed Central Ltd.

 

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

 

Abstract

The stromal vascular fraction (SVF) of adipose tissue is known to contain Mesenchymal stem cells (MSC), T regulatory cells, Endothelial precursor cells, Preadipocytes, as well as anti-inflammatory M2 macrophages. Safety of autologous adipose tissue implantation is supported by extensive use of this procedure in cosmetic surgery, as well as by ongoing studies using in vitro expanded adipose derived MSC. Equine and canine studies demonstrating anti-inflammatory and regenerative effects of non-expanded SVF cells have yielded promising results. Although non-expanded SVF cells have been used successfully in accelerating healing of Crohn's fistulas, to our knowledge clinical use of these cells for systemic immune modulation has not been reported. In this communication we discuss the rationale for use of autologous SVF in treatment of multiple sclerosis and describe our experiences with three patients. Based on this rationale and initial experiences, we propose controlled trials of autologous SVF in various inflammatory conditions.

 

1. Introduction

Adipose tissue has attracted interest as a possible alternative stem cell source to bone marrow. Enticing characteristics of adipose derived cells include: a) ease of extraction, b) higher content of mesenchymal stem cells (MSC) as compared to bone marrow, and c) ex vivo expandability of MSC is approximately equivalent, if not superior to bone marrow [1]. With one exception [2], clinical trials on adipose derived cells, to date, have been limited to ex vivo expanded cells, which share properties with bone marrow derived MSC [3-8]. MSC expanded from adipose tissue are equivalent, if not superior to bone marrow in terms of differentiation ability [9,10], angiogenesis stimulating potential [11], and immune modulatory effects [12]. Given the requirements and potential contaminations associated with ex vivo cellular expansion, a simpler procedure would be the use of primary adipose tissue derived cells for therapy. Indeed it is reported that over 3000 horses with various cartilage and bone injuries have been treated with autologous lipoaspirate fractions without cellular expansion [13]. In double blind studies of canine osteoarthritis statistically significant improvements in lameness, range of motion, and overall quality of life have been described [14,15].

If such approaches could be translated clinically, an easy-to-use autologous stem cell therapy could be implemented that is applicable to a multitude of indications. Indeed, this is the desire of commercial entities that are developing bench top closed systems for autologous adipose cell therapy, such as Cytori's Celution™ system [16] and Tissue Genesis' TGI 1000™ platform [17], which are presently entering clinical trials. Unfortunately, since the majority of scientific studies have focused on in vitro expanded adipose derived cells, relatively little is known about the potential clinical effects of the whole lipoaspirate that contains numerous cell populations besides MSC. From a safety perspective the process of autologous fat grafting has been commonly used in cosmetic surgery [18,19], so at least theoretically, autologous cell therapy, with the numerous cellular populations besides MSC that are found in adipose tissue, should be relatively innocuous. However, from an efficacy or disease-impact perspective, it is important to consider the various cellular components of adipose tissue and to develop a theoretical framework for evaluating activities that these components may mediate when administered systemically. For example, while attention is focused on the MSC component of adipose tissue, the high concentrations of monocytes/macrophages, and potential impact these may have on a clinical indication is often ignored.

In this paper we will discuss the potential use of the adipose derived cells for the treatment of inflammatory conditions in general, with specific emphasis on multiple sclerosis. Due to the chronic nature of the disease, the fact that in some situations remission naturally occurs, as well as lack of therapeutic impact on long term progression of current treatments, we examine the possibility of using autologous adipose derived cells in this condition. We will discuss the cellular components of adipose tissue, the biology of these components, how they may be involved in suppression of inflammatory/immunological aspects of MS, and conclude by providing case reports of three patients treatment with autologous adipose derived cells.

 

2. Components of Adipose Tissue

Mesenchymal Stem CellsMSC

The mononuclear fraction of adipose tissue, referred to as the stromal vascular fraction (SVF) was originally described as a mitotically active source of adipocyte precursors by Hollenberg et al. in 1968 [20]. These cells morphologically resembled fibroblasts and were demonstrated to differentiate into pre-adipocytes and functional adipose tissue in vitro [21]. Although it was suggested that non-adipose differentiation of SVF may occur under specific conditions [22], the notion of "adipose-derived stem cells" was not widely recognized until a seminal paper in 2001, where Zuk et al demonstrated the SVF contains large numbers of mesenchymal stem cells (MSC)-like cells that could be induced to differentiate into adipogenic, chondrogenic, myogenic, and osteogenic lineages [23]. Subsequent to the initial description, the same group reported after in vitro expansion the SVF derived cells had surface marker expression similar to bone marrow derived MSC, comprising of positive for CD29, CD44, CD71, CD90, CD105/SH2, and SH3 and lacking CD31, CD34, and CD45 expression [24]. Boquest et al characterized fresh CD45 negative, CD34 positive, CD105 positive SVF cells based on CD31 expression. They demonstrated that the CD31 negative cells exhibited mesenchymal properties and could be expanded in vitro, whereas the CD31 positive cells possessed endothelial-like properties with poor in vitro expansion capacity [25]. Mesenchymal cells with pluripotent potential have also been isolated from the liposuction aspirate fluid, which is the fluid portion of liposuction aspirates [26].

Endothelial Progenitor CellsEPC

In addition to MSC content, it was identified that SVF contains endothelial precursor cells (EPC). A common notion is that vasculature tissue continually replenishes damaged endothelial cells de novo from circulating bone marrow derived EPC [27], and that administration of exogenous EPC in animals having damaged vasculature can inhibit progression of atherosclerosis or restenosis [28,29]. Miranville et al demonstrated that human SVF cells isolated from subcutaneous or visceral adipose tissue contain a population of cells positive for CD34, CD133 and the drug efflux pump ABCG2 [30]. These cells had endothelial colony forming ability in vitro, and in vivo could induce angiogenesis in a hindlimb ischemia model. Interestingly, the concentrations of cells with the phenotype associated with in vivo angiogenic ability, CD31 negative and CD34 positive, was positively associated with body mass index. This suggests the possibility that endothelial precursor cell entrapment in adipose tissue of obese patients may be related to the reduced angiogenic function seen in obesity [31]. Several other groups have reported CD34 positive cells in the SVF capable of stimulating angiogenesis directly or through release of growth factors such as IGF-1, HGF-1 and VEGF [32-35]. The existence of a CD34 positive subset in the SVF may indicate possibility of cells with not only endothelial but also hematopoietic potential. Indeed at least one report exists of a Bipotent hematopoietic and angiopoietic phenotype isolated from the SVF [36]. Thus from these data it appears that SVF contains at least 2 major populations of stem cells, an MSC compartment and an EPC compartment that may have some hematopoietic activity. When these cells are quantified, one author describes that from primary isolated SVF, approximately 2% of the cells have the hematopoietic-associated CD34+ CD45+ phenotype, and 6.7% having a mesenchymal CD105+ CD146+ phenotype [37]. Many studies using SVF perform in vitro expansion of the cells, this causes selection for certain cell populations such as MSC and decreases the number of CD34 cells [38]. Thus in vitro expanded SVF derived cells can not be compared with primary isolated SVF cells.

Immune Regulatory Monocytes/Macrophages

In addition to its stem/progenitor cell content, the SVF is known to contain monocytes/macrophages. Although pluripotency of monocytic populations has previously been described [39,40], we will focus our discussion to immunological properties. Initial experiments suggested that Macrophage content of adipose tissue was associated with the Chronic low grade inflammation found in Obese patients. This was suggested by co-culture experiments in which adipocytes were capable of inducing TNF-alpha secretion from macrophage cell lines in vitro [41]. Clinical studies demonstrated that adipocytes also directly release a constitutive amount of TNF-alpha and leptin, which are capable of inducing macrophage secretion of inflammatory mediators [42]. It appears from several studies in mice and humans that when Monocytes/Macrophages are isolated from adipose tissue, they in fact possess anti-inflammatory functions characterized by high expression of IL-10 and IL-1 receptor antagonist [43-45]. These adipose derived macrophages have an "M2" phenotype, which physiologically is seen in conditions of immune suppression such as in tumors [46], post-sepsis compensatory anti-inflammatory syndrome [47,48], or pregnancy associated decidual macrophages [49]. It is estimated that the monocytic/macrophage compartment of the SVF is approximately 10% based on CD14 expression [37]. Interestingly, administrations of ex vivo generated M2 macrophages have been demonstrated to inhibit kidney injury in an adriamycin-induced model [50]. In the context of MS, alternatively activated, M2-like microglial cells are believed to inhibit progression in the EAE model [51]. Thus the anti-inflammatory activities of M2 cells are a potential mechanism of therapeutic effect of SVF cells when isolated from primary sources and not expanded.

T Regulatory Cells

It has been reported by us and others, that activation of T cells in the absence of costimulatory signals leads to generation of immune suppressive CD4+ CD25+ T regulatory (Treg) cells [52,53]. Thus local activation of immunity in adipose tissue would theoretically be associated with reduced costimulatory molecule expression by the M2 macrophages, which theoretically may predispose to Treg generation. Conversely, it is known that Tregs are involved in maintaining macrophages in the M2 phenotype [54]. Supporting the possibility of Treg in adipose tissue also comes from the high concentration of local MSC which are known to secrete TGF-beta [55] and IL-10 [56], both involved in Treg generation [57]. Indeed numerous studies have demonstrated the ability of MSC to induce Treg cells [56,58-60]. To test the possibility that Treg exist in the SVF, we performed a series of experiments isolating CD4, CD25 positive cells from the SVF of BALB/c mice and compared frequency between other tissues, (lymph node and spleen). We observed a 3 fold increase in the CD4+, CD25+ compartment as compared to control tissues. Functionally, these cells were capable of suppressing ConA stimulated syngeneic CD4+ CD25+ negative cells (manuscript in preparation).

 

3. Treatment of Autoimmunity with Adipose Cells

In general, MSC, whether derived from the bone marrow, adipose, or other sources, have been demonstrated to exert dual functions that are relevant to autoimmunity [61-65]. These conditions are usually exemplified by activation of innate immune components, breakdown of self tolerance of the adaptive immune response, and subsequent destruction of tissues. Although these are generalizations, an initial insult either by foreign microorganisms, or other means, causes tissue damage and activation of innate immunity, which under proper genetic background leads to re-activation/escape from anergy of "self"-recognizing T cell clones, thus causing more tissue damage, activation of immunity, and lose of function. MSC inhibit innate immune activation by blocking Dendritic cell maturation [66,67], by suppressing Macrophage activation [68], and by producing agents such as IL-1 receptor antagonist [69] and IL-10 [70] that directly block inflammatory signaling. Perhaps the strongest example of MSC inhibiting the innate immune response is the recent publication of Nemeth et al, which demonstrated that administration of MSC can block onset of sepsis in the aggressive cecal ligation and puncture model [68]. Through inhibiting DC activation, MSC suppress subsequent adaptive immunity by generating T regulatory (Treg) cells [59], as well as blocking cytotoxic activities of CD8 cells. In some situations, increased immunoregulatory activity is reported with expanded MSC compartment of SVF as reported by Mcintosh et al. [71].

In addition to inhibiting pathological innate and adaptive immunity, MSC have the ability to selectively home to areas of tissue damage, and mediate direct or indirect repair function. As an example, CXCR-4 expression of MSC allows homing toward injured/hypoxic tissue after intravenous administration. Indeed this has allowed for numerous studies demonstrating positive effects of intravenously administered MSC causing regeneration in many tissues such as CNS injury [72,73], transplant rejection [59], toxin-induced diabetes [74], nephropathy [75], and enteropathy [76]. The regenerative effects of MSC have been postulated to be mediated by differentiation into damaged tissue, although this is somewhat controversial, as well as through secretion of growth factors/antiapoptotic factors which induce tissue regeneration [77,78].

The ability of MSC to inhibit immune response, while offering the possibility of inducing/accelerating healing of tissue that has already been damaged, makes this population attractive for treatment of autoimmune disorders. While numerous studies clinical studies are using expanded MSC derived from the bone marrow [79-81], here we chose an indication of autologous adipose SVF based on the immunological profile, the length of disease progress allowing several interventions, and the fact that the disease naturally has periods of remission during which the rationale would be to amplify a process that already is underway.

 

4. Multiple Sclerosis

Multiple sclerosis (MS) is an autoimmune condition in which the immune system attacks the central nervous system (CNS), leading to demyelination. It may cause numerous physical and mental symptoms, and often progresses to physical and cognitive disability. Disease onset usually occurs in young adults, and is more common in women [82]. MS affects the areas of the brain and spinal cord known as the white matter. Specifically, MS destroys oligodendrocytes, which are the cells responsible for creating and maintaining the myelin sheath, which helps the neurons carry electrical signals. MS results in a thinning or complete loss of myelin and, less frequently, transection of axons [83].

Current therapies for MS include steroids, immune suppressants (cyclosporine, azathioprine, methotrexate), immune modulators (interferons, glatiramer acetate), and immune modulating antibodies (natalizumab). At present none of the MS treatment available on the market selectively inhibit the immune attack against the nervous system, nor do they stimulate regeneration of previously damaged tissue.

 

Treg cells modulate MS

Induction of remission in MS has been associated with stimulation of T regulatory cells. For example, patients responding to the clinically used immune modulatory drug glatiramer acetate have been reported to have increased levels of CD4+, CD25+, FoxP3+ Treg cells in peripheral blood and cerebral spinal fluid [84]. Interferon beta, another clinically used drug for MS induces a renormalization of Treg activity after initiation of therapy through stimulation of de novo regulatory cell generation [85]. In the animal model of MS, experimental allergic encephalomyelitis (EAE), disease progression is exacerbated by Treg depletion [86], and natural protection against disease in certain models of EAE is associated with antigen-specific Treg [87]. Thus there is some reason to believe that stimulation of the Treg compartment may be therapeutically beneficial in MS.

 

Endogenous neural stem cells affect MS recovery

In addition to immune damage, MS patients are known to have a certain degree of recovery based on endogenous repair processes. Pregnancy associated MS remission has been demonstrated to be associated with increased white matter plasticity and oligodendrocyte repair activity [88]. Functional MRI (fMRI) studies have suggested that various behavioral modifications may augment repair processes at least in a subset of MS patients [89]. Endogenous stem cells in the sub-ventricular zone of brains of mice and humans with MS have been demonstrated to possess ability to differentiate into oligodendrocytes and to some extent assist in remyelination [89]. For example, an 8-fold increase in de novo differentiating sub-ventricular zone derived cells was observed in autopsy samples of MS patients in active as compared to non-active lesions [90].

 

Stem Cell Therapy for MS

The therapeutic effects of MSC in MS have been demonstrated in several animal studies. In one of the first studies of immune modulation, Zappia et al. demonstrated administration of MSC subsequent to immunization with encephalomyelitis-inducing bovine myelin prevented onset of the mouse MS-like disease EAE. The investigators attributed the therapeutic effects to stimulation of Treg cells, deviation of cytokine profile, and apoptosis of activated T cells [73]. It is interesting to note that the MSC were injected intravenously. Several other studies have shown inhibition of EAE using various MSC injection protocols [91,92].

To our knowledge there is only one publication describing clinical exploration of MSC in MS. An Iranian group reported using intrathecal injections of autologous culture expanded MSC in treatment unresponsive MS patients demonstrated improvement in one patient (EDSS score from 5 to 2.5), no change in 4 patients, and progressive disease in 5 patients based on EDSS score. Functional system assessment revealed six patients had improvement in their sensory, pyramidal, and cerebellar functions. One showed no difference in clinical assessment and three deteriorated [93].

 

5. Case Reports

Given the rationale that autologous SVF cells have a reasonable safety profile, and contain both immune modulatory and regenerative cell populations, a physician-initiated compassionate-use treatment was explored in 3 patients. Here we describe their treatments and histories.

#CR-231

In 2005, a 50-year-old man was diagnosed with Relapsing-remitting MS, presenting with tonic spasms, stiffness, gait imbalance, excessive hearing loss, loss of coordination, numbness in both feet, sexual dysfunction, severe pain all over his body, fatigue and depression. In 2005, the patient experienced refractory spells of tonic flexion spasms, occurring for several minutes at a time and multiple times throughout the day. He was treated with muscle relaxants, I.V. steroids and Tegretol, and his condition had improved. However, in 2006 he experienced severe uncontrollable tonic extensions of all four extremities lasting about two minutes and associated with significant pain. Cranial MRI done at that time revealed at least 30 periventricular white matter lesions. Patient also reported excellent response to Solu-Medrol infusions. Therefore, the combination of response to steroids, characteristic MRI abnormalities and positive oligoclonal banding strongly suggested a diagnosis of Relapsing Remitting MS. Infusions of Tysabri (Natalizumab, Biogen Idec) every four weeks were prescribed in November 2006, with excellent results and no significant side effects. However, in March 2007 patient reported spasticity approximately three weeks after the infusions, leading to alteration of his Tysabri infusion regimen to Q3 weeks. By June 2007 the patient had began complaining of significant memory loss and by September 2007 he has had recurrence of his tonic spasms with multiple attacks daily. He was treated with Solu-Medrol, Baclofen, Provigil, Tegretol, Trileptal, Tysabri, Vitamins, Omega-3 and Zanaflex with some improvement of his neurologic symptoms. However, he complained of severe abdominal pain, decreased appetite and melanotic stools, consistent with stress ulcer secondary to steroid treatment. By November 2007 the patient was still somewhat responsive to Tysabri and I.V. Solu-Medrol, but continued to experience multiple severe tonic spasms at a rate of 30 – 40 spasms per month.

In May 2008, the patient was treated with two I.V. infusions of 28 million SVF cells and multiple intrathecal and intravenous infusions of allogeneic CD34+ and MSC cells. MSC were third party unmatched and CD34 were matched by mixed lymphocyte reaction. Infusions were performed within a 9-day period and were very well tolerated without any adverse or side effects. No other treatments were necessary during the patient's stay. After the second stem cell infusion the patient reported a significant decrease of his generalized pain. However, he continued to experience severe neck and shoulder pain and was re-evaluated by his neurologist. Two months after the stem cell therapy, the volume of his hearing aids had to be lowered once per week over 4 weeks. Three months after the stem cell infusions the patient reported a significant improvement of his cognition and almost complete reduction of the spasticity in his extremities. He mentioned that he has had 623 tonic seizures in the past and confirmed that he has not experienced any more seizures since the completion of the stem cell therapy. A neurological evaluation performed three months after the stem cell infusions revealed an intact cranial nerve (II-XII) function and no nystagmus, normal motor function without any atrophy or fasciculations, and intact sensory and cerebellar functions and mental status. New MRI images, obtained 6 months after the stem cell treatment showed lesions, very similar to the lesions observed before the stem cell treatment (Figure 1). The patient also reported significantly improved memory, sexual function, and energy level. Currently, the patient is taking only multivitamin, minerals and Omega 3.

 

Figure 1. MRI Images obtained before (Panels A and B), and six months after (Panel C) the stem cell treatment of patient 1. Panels A and B: Consecutive axial FLuid-Attenuated Inversion Recovery (FLAIR) images through the lateral ventricles show multiple small foci of bright signal in the periventricular and subcortical white matter, consistent with plaques of multiple sclerosis. Panel C: Axial FLAIR image shows no significant change in the multiple periventricular and subcortical white-matter plaques. (For the comparison, note that this slice is positioned between those in A and B, and at slightly different scanning-angle, so it includes lesions of both those slices, as well as others slightly out-of their plane.).

Charlie 發表在 痞客邦 留言(0) 人氣()

1496276260001104659-10-026386_G95952BG09I0111321465840155_blogHeader_promethean  

Autologous stromal vascular fraction therapy for Rheumatoid arthritisrationale and clinical safety

Jorge Paz Rodriguez1, Michael P Murphy2, Soonjun Hong2, Marialaura Madrigal1, Keith L March2, Boris Minev3, Robert J Harman4, Chien-Shing Chen5, Ruben Berrocal Timmons6, Annette M Marleau7* and Neil H Riordan1

*Corresponding author : Annette M Marleau amarleau@immed.org

Author Affiliations

1 Medistem Panama, Panama City, Panama

2 Indiana University, Indiana, USA

3 University of San Diego, San Diego, CA, USA

4 Vet-Stem Inc, Poway, CA, USA

5 Division of Hematology and Oncology, Loma Linda University, School of Medicine, Loma Linda, CA, USA

6 Senacyt, Panama City, Panama, USA

7 Institute for Molecular Medicine, Huntington Beach, CA, USA

 

International Archives of Medicine 2012, 5:5

doi:10.1186/1755-7682-5-5

 

The electronic version of this article is the complete one and can be found online at: http://www.intarchmed.com/content/5/1/5

Received : 8 December 2011

Accepted : 8 February 2012

Published : 8 February 2012

© 2012 Paz Rodriguez et al; licensee BioMed Central Ltd.

 

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

 

Abstract

Advancements in rheumatoid arthritis (RA) treatment protocols and introduction of targeted biological therapies have markedly improved patient outcomes, despite this, up to 50% of patients still fail to achieve a significant clinical response. In veterinary medicine, stem cell therapy in the form of autologous stromal vascular fraction (SVF) is an accepted therapeutic modality for degenerative conditions with 80% improvement and no serious treatment associated adverse events reported. Clinical translation of SVF therapy relies on confirmation of veterinary findings in targeted patient populations. Here we describe the rationale and preclinical data supporting the use of autologous SVF in treatment of RA, as well as provide 1, 3, 6, and 13 month safety outcomes in 13 RA patients treated with this approach.

 

Introduction

Increasing number of reports support the possibility of utilizing adult stem cell therapy not only for treatment of degenerative conditions, but also as a means of addressing underlying inflammation or autoimmune conditions [1-8]. Unfortunately, stem cell therapy is often complicated by the need for complex laboratories, processing procedures and clean rooms. The potential drawbacks of allogeneic donor approaches include the possibility of eventual rejection of the cellular graft [9-12], as well as limitation of efficacy due to trophic effects but not de novo tissue generation [13-15]. Conversely, adult stem cell based approaches, particularly using bone marrow, are limited to the relatively small number of progenitor cells within the bone marrow. While bone marrow mononuclear cell administration appears to be effective in conditions where cells are locally implanted, such as intramyocardial [16-18], or intramuscular in critical limb ischemia [19-21], the intravenous administration of non-expanded bone marrow has not been performed with efficacy in systemic conditions without prior myeloablation of the recipient. One way of circumventing this problem is to expand autologous stem cells prior to implantation. Unfortunately, besides issues of cost and practicality, there is a risk that the in vitro manipulation could be linked to contamination, as well as genomic alterations of the cells, leading to transformation.

Several studies have used bone marrow derived mesenchymal stem cells (MSC) for various conditions including type 2 diabetes [22], osteoarthritis [23], stroke [24], and amyotrophic lateral sclerosis [25]. This procedure requires expansion of the MSC compartment in vitro and therefore adds an element of complexity to the treatment. A much simpler procedure, for which adipose tissue is uniquely suited, is the administration of autologous, non-expanded cellular fraction. The rationale behind this derives from observations that: a) adipose tissue contains substantially higher numbers of MSC compared to bone marrow [26]; b) MSC from adipose tissue do not appear to decrease in number as a result of age [27,28]; and c) adipose tissue contains unique populations of cells including high concentrations of Endothelial progenitor cells and T regulatory (Treg) cells that express up to 100-fold higher levels of the immune suppressive cytokine IL-10 as compared to circulating Tregs [29].

The adipose stromal vascular fraction (SVF) is comprised of the Mononuclear cells derived from adipose tissue. This term is more than 4 decades old, used to describe the mitotically active source of adipocyte precursors [30,31]. SVF as a source of stem cells was first described by Zuk et al. who identified MSC-like cells in SVF that could be induced to differentiate into Adipogenic, Chondrogenic, Myogenic, and Osteogenic lineages [32]. Subsequent to the initial description, the same group reported after in vitro expansion, the SVF derived cells had surface marker expression similar to bone marrow derived MSC, comprising CD29, CD44, CD71, CD90, CD105/SH2, and SH3 and lacking CD31, CD34, and CD45 expression [33,34].

Reported clinical trials on adipose derived cells, to date, have all utilized ex vivo expanded cells, which share properties with bone marrow derived MSC [35-40]. MSC expanded from adipose tissue are equivalent, if not superior to bone marrow in terms of differentiation ability [41,42], angiogenesis stimulating potential [43,44], and immune modulatory effects [45]. Given the requirements and potential contaminations associated with ex vivo cellular expansion, a simpler procedure would be the use of primary adipose tissue derived cells for therapy. Indeed, it is reported that over 4000 horses and 4000 dogs with various cartilage and bone injuries have been successfully treated with autologous SVF [46]. In double blind studies of canine osteoarthritis statistically significant improvements in lameness, range of motion, and overall quality of life have been described [47,48].

If such approaches could be translated clinically, an easy-to-use autologous stem cell therapy could be implemented that is applicable to a multitude of indications. Indeed, this is the desire of commercial entities that are developing bench-top closed systems for autologous adipose cell therapy [49,50], which are presently entering clinical trials. Unfortunately, since the majority of scientific studies have focused on in vitro expanded adipose derived cells, relatively little is known about the potential clinical effects of the whole lipoaspirate that contains numerous cell populations besides MSC. From a safety perspective the process of autologous fat grafting has been commonly used in cosmetic surgery [51,52]. Therefore, administration of autologous heterogeneous adipose cellular fractions, which contain numerous cellular populations besides MSC, should be relatively innocuous. However, from an efficacy or disease-impact perspective, it is important to consider the various cellular components of adipose tissue and to develop a theoretical framework for evaluating activities that these components may mediate when administered systemically. For example, while attention is focused on the MSC component of adipose tissue, the high concentrations of Monocytes/Macrophages, and potential impact these may have on a clinical indication is often ignored [29].

In the published literature, the clinical use of systemically administered SVF cells has been reported in two pilot studies by our group. The first was a description of 3 patients suffering from multiple sclerosis who received intravenous administration of autologous adipose SVF as part of a cellular cocktail. All 3 patients reported significant improvement neurologically, and demonstrated an excellent safety profile [53]. Additionally, a 1 patient case report described a remission of RA subsequent to administration of autologous SVF as a monotherapy [29]. Here we will provide a description of adipose SVF components, provide a rationale for use, and describe safety at 1, 3, 6, and 12 months in a 13 patient retrospective analysis.

 

The MSC component of adipose tissue

MSC are conventionally extracted from bone marrow sources as a cellular therapy for inflammatory associated conditions. Specifically, the most advanced clinical trials in the area of regenerative medicine have been performed by the company Osiris, whose main product is a "universal donor" MSC, termed "Prochymal"[54]. This cellular product has entered Phase III trials in graft versus host disease, and is currently being tested for heart failure [55]. Other bone marrow derived MSC-like products are in clinical trials, for example, Mesoblast is in Phase III assessing its Mesenchymal Precursor Cell for efficacy in post hematopoietic transplant graft failure, as well as in Phase II for heart failure [56]. Therapeutic advantages of MSC include their ability to migrate to injured tissue, in part via detections of hypoxia through the CXCR4-SDF-1 axis [57,58], differentiation activity into multiple tissues [59,60], release of trophic factors [61], inhibition of apoptosis [62-64], stimulation of angiogenesis [65], inhibition of inflammation [66], and stimulation of Treg activity [67]. Despite the advantages of the current approaches, bone marrow contains relatively small numbers of MSC, thus, as previously mentioned, therapeutics with bone marrow for systemic applications requires ex vivo expansion. Specifically, the bone marrow contains approximately 1/10,000 to 1/100,000 MSC per nucleated cells [68], whereas adipose tissue contains approximately 1001000 fold higher MSC concentration, or approximately 50100,000 MSC per mL [69]. Given the relative ease of extracting 500 mL of lipoaspirate, it is conceptually feasible to generate a 2550 million cell dose of MSC, which is close to the systemic doses of MSC that are typically used in clinical trials of allogeneic expanded cells (eg. 50-100 million cells in various clinical trials) [34]. Conceptually, given that the MSC present in the SVF are autologous, one could envision higher therapeutic potential due to the lack of allo-immune clearance as compared to allogeneic MSC, although this needs to be assessed experimentally.

Adipose MSC contain several similarities and differences as compared to bone marrow derived MSC, although this area is still considered to be controversial. Specifically, in animal cardiac infarct models it has been demonstrated that that expanded adipose MSC are superior to bone marrow MSC in terms of stimulating angiogenesis, decreasing cardiac pathology, and stimulating VEGF and FGF secretion [70]. Using an in vivo lentiviral-labeled system, it was demonstrated that adipose derived MSC have a superior ability to BM derived MSC to integrate into cardiac muscle after injury, as well as to restore function [71]. In addition to specific propensities for differentiation, adipose tissue-derived MSC appear to be superior to bone marrow in terms of proliferative potential without loss of Telomere length. Vidal et al. demonstrated that adipose MSC could multiply for almost twice as many cell passages without undergoing senescence as compared to bone marrow MSC [72].

Conversely, several authors have reported similarities between bone marrow and SVF MSC sources. For example, subsequent to exposure to chemotactic agents, both sources were reported to yield MSC possessing similar rates of migration [73]. The same study also demonstrated comparable ability to generate cartilage when treated under differentiation conditions. Another study reported exposure of bone marrow or adipose derived MSC to Ischemia leads to the release of similar levels of angiogenic factors, as well as resistance to apoptosis when cultured in Hypoxic environments [74]. Comparison of immunological properties led to the conclusion that when expanded, both BM and adipose derived MSC appear to have similar properties in terms of suppressing mixed lymphocyte reactions, inhibiting release of type 1 and inflammatory cytokines, as well as generating progeny cells that appear to be relatively immune privileged [75]. These data were confirmed by the group of Zhang et al. who compared cord blood, bone marrow, and adipose MSC and found almost identical ability to inhibit immune response assays in vitro [76]. In contrast, Najar et al. reported that Wharton Jelly and Adipose derived MSC were superior immune suppressors as compared to BM MSC in terms of inhibiting lymphocyte proliferation and type 1 cytokine production [77,78]. An important consideration is that there is a great deal of variability between studies, not only in the tissue sources from which MSC are derived but also in terms of cell isolation, culture and expansion methods as well as donor-specific characteristics that could conceivably influence the activities and differentiation potential of these cells [79]. Therefore, one potential disadvantage of utilizing ex vivo-manipulated MSC is the potential for introducing more heterogeneity in their regenerative capabilities. For treatments involving autologous MSC, patient-to-patient differences in MSC function could also lead to variability in the clinical efficacy of treatments.

Clinically, adipose derived MSC have been used in treatment of 8 spinal cord injury patients in Korea where administration of autologous expanded MSC at doses of 400 million per patient did not elicit treatment associated adverse events during a 3-month follow-up [80]. Additionally, this study also reported genetic stability of MSC in vitro and lack of toxicity or tumorigenicity of MSC in immune deficient mice. Trivedi's group treated 11 patients with type 1 diabetes using a combination of autologous adipose derived MSC that were cultured in a pro-pancreatic differentiation media together with cultured bone marrow MSC. No adverse effects were noted over an average of 23 months follow-up period and a decrease in insulin requirements was noted [81]. Garcia-Olmo et al. reported a study where autologous expanded adipose MSC were administered to patients with complex perianal fistulas, with 35 of cryptoglandular origin and 14 associated with Crohn's disease. They observed that fistula healing occurred in 17 (71 percent) of 24 patients who received ASCs in addition to fibrin glue compared with 4 (16 percent) of 25 patients who received fibrin glue alone (relative risk for healing, 4.43; confidence interval, 1.74-11.27; P < 0.001). The proportion of patients with documented fistula healing was similar in Crohn's and non-Crohn's subgroups. ASCs were also more effective than fibrin glue alone in patients with a suprasphincteric fistulous tract (P = 0.001). Furthermore, quality of life scores were higher in patients who received ASCs than in those who received fibrin glue alone [82]. Due to the anti-inflammatory effects of MSC in general [83-86], and specifically the ability of MSC to inhibit graft versus host disease (GVHD) [87,88], Fang et al. reported a series of pilot cases in which patients with steroid refractory GVHD was successfully treated by administration of autologous adipose derived expanded MSC [89,90].

Thus it appears that the MSC component of adipose tissue possesses numerous preclinical and clinical therapeutic properties and may be an important component of the SVF cell population that is responsible for therapeutic effects observed after administration.

 

Adipose tissue resident T regulatory (Treg) cells

Treg cells are conventionally described as CD4+ cells possessing the transcription factor FoxP3 and capable of suppressing T cell activation, dendritic cell maturation, neutrophil activation, and antibody production. The fundamental role of Treg in controlling immunity can be illustrated by the fact that genetic mutations associated with loss of Treg function, such as FoxP3 mutations, are associated with autoimmunity in mouse and man [91-93]. Additionally, conditional ablation of the Treg compartment in genetically-engineered mice results in systemic organ autoimmunity [94]. Numerous autoimmune conditions enter remission as a result of increased Treg number and/or activity, whereas relapse is associated with reduction in number and/or activity. Specifically, this has been demonstrated in multiple sclerosis [95-99], rheumatoid arthritis [100-104], and lupus [105-107]. Given the importance of Treg cells in the control of autoimmunity, it would be useful to possess sources of Treg cells that are easily accessible and can be reintroduced into the patient for immune modulation. It has been previously demonstrated that high numbers of Treg cells are found in adipose tissue at concentrations much higher than other peripheral compartments such as blood or spleen [108]. Interestingly, adipose derived Treg contain approximately 100 fold higher concentrations of the immune regulatory effector cytokine IL-10 [109,110]. It is known that the adipose derived cytokines Leptin and TNF-alpha inhibit Treg proliferation and activity in vivo [111,112]. The local effects of these cytokines would conceptually, be negated by liberating Treg from fat tissue followed by systemic re-administration, resulting in enhanced Treg activity. Administration of a large number of Treg cells with augmented in vivo proliferative and functional potential may result in a reduction of the threshold needed to attain tolerance to an ongoing immune response. For example, anti-CD3 antibodies have been reported to induce antigen-specific tolerance, despite the fact that the surge in Treg numbers was not antigen-specific [113]. Thus one conceptual advantage of utilizing SVF therapy would be not only the MSC content, which possesses various regenerative properties, but also Treg, which would enhance anti-inflammatory/tolerance inducing properties. Given that both MSC and Treg are considered to be tolerance-promoting, it may be feasible to consider that synergize of tolerance induction may be occurring when the two cell populations are co-administered in the form of SVF.

 

Endothelial progenitor cell (EPC)

Aging and/or damaged blood vessel endothelium is constantly renewed by circulating cells termed endothelial progenitor cells (EPC). This notion gathered significant scientific following subsequent to a paper by Asahara et al. who demonstrated that BM-originating cells expressing VEGFR-2 and CD34 are capable of incorporating into sites of angiogenesis induced by wire injury or ischemia [114]. Therapeutic properties of EPC have been demonstrated in that administration of exogenous EPC increases vascular repair. This has been shown using in vitro generated EPC, or bone marrow as a source of EPC in myocardial infarct [115,116], stroke [117], lung injury [118-120], liver failure [121-123], and endothelial injury atherosclerotic models [124,125]. Furthermore, administration of growth factors that stimulate mobilization of bone marrow stem cells and EPC have demonstrated therapeutic benefit in animal models of ischemic disease [126,127] as well as endothelial damage [128]. Clinical trials using EPC or bone marrow as a source of EPC for cardiovascular conditions [129-132], have demonstrated some therapeutic benefit, although work is ongoing.

Historically, the bone marrow has been used as a source of EPC, however, numerous recent studies have demonstrated a high content of EPC in adipose tissue [133,134]. Functional demonstration of adipose EPC was performed in experiments in which CD34 expressing cells were sorted for from SVF. This cellular fraction was demonstrated to induce angiogenesis in immune compromised mice that were subjected to hindlimb ischemia. Mechanistically, the cells were identified as EPC based on ability to form endothelial colonies when cultured in vitro [135]. Numerous groups have reported that SVF contains cellular activity that stimulates angiogenesis, for example, Sumi et al. showed that administration of SVF but not adipocytes led to revascularization in the hindlimb ischemia model [136]. Other studies have shown that not only are EPC-like activities found in SVF [137], but also that conditioned media from SVF is capable of stimulating host angiogenesis [138,139]. It is reported that EPC in the SVF stimulate angiogenesis directly through Differentiating into endothelial cells or through Release of growth factors such as IGF-1, HGF-1 and VEGF [136,137,139,140]. Although back-to-back comparisons of bone marrow and adipose derived EPC for assessing angiogenic potential have not been performed, the substantially higher concentration of these cells in SVF supports the investigation of this tissue as a practical cell source for clinical applications.

 

Rationale for clinical applications

Given that SVF represents a multi-cellular population containing MSC, Treg, and EPC, the potential for therapeutic utilization would include many conditions that require regeneration, immune modulation, and possibly angiogenesis.

We previously reported remission in a patient with rheumatoid arthritis who was treated with autologous SVF [29]. Animal studies using the collagen II model of RA have demonstrated that administration of MSC is associated with immune modulation [141-143], disease remission [144,145], and regeneration of cartilage [5,146]. Additionally, our group and others have reported that Treg cells are associated with induction of disease remission [100-102,104,147-151].

 

Safety data

The study was a retrospective analysis of patients treated under the practice of medicine under doctor patient privilege. The protocols were approved by local and institutional committees and all patients signed informed consent forms explaining the unproven and experimental nature of the treatment. Retrospective chart analysis of the patients was approved by PEARL IRB (Indianapolis, Indiana).

Patients received the indicated amount of cells by intravenous injection (2x106 cells per ml diluted in Saline solution), intra-articular injection (2.5x106 cells per ml in each injured joint, diluted in Saline solution and the patient's own serum). Multiple injections of cells were given to increase the therapeutic efficacy. Follow-ups were performed for all patients at 1, 3, 6 and 12 months.

SVF cells were isolated and prepared under the guidelines of Good Tissue Practices 21 CFR 1271 as relates to sample screening and processing in the sterile flow hood, inside of a class 10000 clean room. SVF cells were isolated by first washing 500 cc of Lipoaspirate with PBS and subsequently, the cells were transferred to 175 ml sterile centrifuge containers followed by the addition of Collagenase solution for a final concentration of 0.048%. The centrifuge containers were sealed and placed in an elliptical shaker and incubated at 37 C for 6080 minutes. The content of the tubes was filtered through a cell strainer into sterile 50 ml centrifuge tubes and centrifuged for 12 min at 800 rcf. During centrifugation, SVF cells formed a pellet in the bottom of the container while the adipocyte layer and debris remained suspended. Following centrifugation, the stromal cells were resuspended in 5 mL of autologous serum for enzyme inactivation then washed 2 times with PBS. The fraction used for Intraarticular injection was Incubated with Buffer to lyse red blood cells and Washed once more. All the cells were aliquoted in cryovials, frozen in liquid nitrogen and stored until use. Cells were assessed for viability, endotoxin, and contamination before treatment was performed. The patient was allowed to heal from the liposuction for one week. For each treatment session, after thawing the cells were rinsed with PBS and Human AB serum, diluted in saline solution and autologous serum, loaded into sterile syringes, and then transported in a controlled temperature cooler accompanied by the corresponding certificate and delivered to the physician for infusion.

Thirteen patients with Rheumatoid arthritis were treated with 38148 million SVF cells intravenously and intra-articularly (Table 1). Although no hematopoietic or biological abnormalities were noted, one of the patients reported facial flushing, fever and myalgia after a third of four injections. These symptoms all resolved spontaneously.

 

Table 1. Patient Treatments and Safety Outcome

 

Conclusion

These data suggest the safety and feasibility of administering adipose SVF intravenously. The uses of adipose stem cells have been reported in conditions as diverse as from hearing loss [152], to heart failure [153]. Given the anti-inflammatory, differentiation ability, and trophic factor production by SVF, we are hopeful that these safety data will support ongoing investigation into this novel and easy to access cell population.

 

Competing interests

NHR and JPR are shareholders of Medistem Panama and Medistem Inc. None of the other authors have any competing interests.

 

Authors' contributions

JPR, MPM, SH, MM, KLM, BM, RJH, CC, RBT, AMM, and NHR performed literature review and wrote the manuscript. SH collected and analyzed patient charts. JPR reported on the clinical cases. NHR, conceived the study and rationale for use of SVF in autoimmunity. All authors read and approved the final manuscript.

Charlie 發表在 痞客邦 留言(0) 人氣()

1390217573-1844674495_n1390316183-2934261841390316183-531698202_n1390316183-1718704163_nadult-mesenchymal-stem-cells  

Nonexpanded mesenchymal stem cells for regenerative medicine : yield in stromal vascular fraction from adipose tissues.

Faustini M1, Bucco M, Chlapanidas T, Lucconi G, Marazzi M, Tosca MC, Gaetani P, Klinger M, Villani S, Ferretti VV, Vigo D, Torre ML.

1Dipartimento di Scienze e Tecnologie Veterinarie, per la Sicurezza Alimentare, Università di Milano, Milan, Italy. massimo.faustini@unimi.it

Tissue Eng Part C Methods. 2010 Dec;16(6):1515-21.

doi: 10.1089/ten.TEC.2010.0214.

Epub 2010 Sep 6.

 

Abstract

The adipose-derived stromal vascular fraction (SVF) represents a rich source of mesenchymal cells, potentially able to differentiate into adipocytes, chondrocytes, osteoblasts, myocytes, cardiomyocytes, hepatocytes, and neuronal, epithelial, and endothelial cells. These cells are ideal candidates for use in regenerative medicine, tissue engineering, including gene therapy, and cell replacement cancer therapies. In this work, we aimed to the optimization of the adipose SVF-based therapy, and the effect of the collection site, surgical procedure, and tissue processing techniques on SVF yield was evaluated in terms of cell recovery and live cells, taking into account the effect of gender, age, and body mass index. Adipose tissue samples were recovered from 125 informed subjects (37 males and 88 females; mean age: 51.31 years; range: 15-87 years), and digested in different condition with collagenase. A multivariate linear model put in evidence that in males the best collection site in terms of yield is located in the abdomen, whereas in females the biopsy region do not influence cell recovery; the collection technique, the age, and the body mass index of donor seem not to influence the cell yield. The tissue-processing procedures strongly modify the yield and the vitality of cells : a collagenase concentration of 0.2% and a digestion time of 1 h could be chosen as the best operating conditions.

Charlie 發表在 痞客邦 留言(0) 人氣()

新萬能細胞STAP 被疑研究造假

自由時報 – 2014218〔編譯林翠儀/綜合報導〕

AJ201402060055M

日本理化學研究所團隊成功開發出新型萬能細胞「STAP」,號稱製作方法比iPS細胞(誘導性多功能幹細胞)更簡單且不易癌化,研究報告上月底發表在英國「自然」期刊電子版後,引起全球矚目,但近日網路出現質疑聲浪,指稱論文提供的影像資料不自然。理研17日宣布已對此展開調查,但認為應不致推翻這項研究成果。

1391046567-3096625569

開發出STAP細胞的理研團隊,是由來自美國哈佛大學、日本山梨大學等名校的學者組成,召集人為現年30歲的美女科學家小保方晴子。研究團隊成功地從實驗鼠脾臟內採集淋巴細胞,加入弱酸性溶液刺激後,在27天內培養出STAP細胞,這種新型萬能細胞可變成各種細胞。

AJ201402030078M

以往學界認為,體細胞一旦角色確定後,光是靠外部刺激不可能產生初期化,但STAP細胞的開發不僅打破這項生命科學的常識,而且製作時間較短、不易癌化病變,因此被視為將是ES細胞(胚胎幹細胞)及iPS細胞之後的「第3種萬能細胞」,連iPS細胞之父、日本諾貝爾獎得主山中伸彌也肯定這是一項重要研究成果。

不過,STAP細胞論文發表後,網路卻傳出質疑聲浪,指論文提供的部分影像資料,和過去發表的影像雷同,還有以STAP細胞培養的實驗鼠胎兒相片疑似重複使用,以及影像中有不自然的線條,疑似曾被加工過等。

Charlie 發表在 痞客邦 留言(0) 人氣()

骨髓移植面容老化?40歲婦像70

作者:廖宗慶 | TVBS – 2014217

 20140217190534509

20140217190538461

您有聽過再生不良性貧血再生障礙性貧血(Aplastic anemiaAA)嗎?這是一種骨髓造血細胞減少,造成紅血球、白血球和血小板都會降低的疾病,嚴重可能喪命,不過桃園有一位陳小姐,接受林口長庚的建議,接受造血幹細胞移植,沒想到去年突然出現排斥現象,而後嚴重老化,原本40歲,現在看起來卻像6070歲.全身皮膚乾裂,眼睛、牙齒通通退化。

250px-Aplastic_anemia

d153206

陳小姐:「我做了這個骨髓移植之後,我都不想活了,多給我5年我也不要。」

乾裂佈滿皺紋的臉龐,哭訴悲哀,看似老婦的陳小姐,卻擁有清亮嗓音,因為她其實今年才40歲。

記者:「會抖啦?」

陳小姐:「對,手沒辦法伸直,然後指甲會長這樣,這個淚腺好像阻塞,一直會自己掉眼淚出來。」

populace_info04_pic001

當美容師的陳小姐,2012年時,還是皮膚白皙、保養得宜,但如今全身上下迅速老化,如同6070歲的老婦人,原來陳小姐之前,診斷出罹患再生不良性貧血,結果醫生發現她妹妹的骨髓與她配對相符,建議做造血幹細胞移植。

陳小姐:「那我說不做移植勒,他醫師說會死。」

醫生說,不做會死,沒想到20127月動了手術,3個月後被告知失敗,去年8月開始急速老化。

陳小姐:「希望怎樣,就是他幫我醫好,還我原來的樣子。」

走路、行動都成困難,想到其他地方治療,也被拒絕,林口長庚發表聲明,強調雖然基因配對相合,但移植後還是嚴重排斥,承諾將持續提供治療照護。

p4_3_13

只是再生不良性貧血,也就是骨髓不能生產足夠或新的細胞,來補充血液細胞,包括紅血球、白血球、血小板都會不足,其他醫院醫師表示,嚴重患者如果不治療,致死率幾乎百分之百,而就算移植,的確可能出現排斥現象。

aplastic anemia_213446_lg

5225-19995-1-PB

血液腫瘤科醫師戴承正:「排斥,譬如說在皮膚上,就會產生大量脫屑的現象,那膽道呢,甚至會有膽道閉鎖,如果是骨隨移植,所造成的排斥現象的話,其實對生命,嚴重來講是有致命性的。」

當初移植想救命,如今陳小姐卻說自己生不如死,就怕再治療,也無法回到過往。

qdxdjz

Charlie 發表在 痞客邦 留言(0) 人氣()

2192-18746_4aadf1a0-e30c-406b-8cf6-c2b04fd5c4edDas_schema2

Diapositiva Cell Metabolism

F1.largeF2.largefgf21Figure1Irisin_Villarroya_2012irisin-and-biegenm.3058-F1

冷天發抖10分鐘 相當於運動1小時肌肉組織釋放「鳶尾素」(Irisin)和FGF21促使體內負責儲存熱量的「白脂肪」加入產生熱能行列消耗囤積的脂肪

TVBS – 201425

 

您知道天氣冷也能夠幫助你減肥嗎,澳洲研究發現,冷天氣能夠幫助人體燃燒脂肪,只要冷到發抖,10分鐘所燃燒的脂肪,相當於運動1小時,因為人發抖的時候,肌肉組織會釋放2種荷爾蒙,幫助體內脂肪燃燒來產生熱量。

佈滿白雪的街頭,路人包的緊緊,冷吱吱的天氣,竟然能幫助減肥,澳洲大學學者研究發現,人體只要因為冷到發抖10分鐘,所燃燒的卡路里,相當於運動1個小時。學者:「有事實證明說,如果你的體溫低於華氏98.6度、攝氏37度,你的身體必須要更努力保持體溫,這會讓你的身體增加代謝,來燃燒更多的熱量。」

研究發現,只要在低溫15C,人體就會開始發抖,肌肉組織會釋放「鳶尾素」(Irisin)和FGF21兩種荷爾蒙,促使體內負責儲存熱量的「白脂肪」,加入產生熱能行列,來消耗囤積的脂肪。新聞主播:「天冷不會讓你減少很多體重,有可能是燃燒掉一些卡路里,但是如果你把它當成是冬天減肥的唯一方法,那你就去試吧。」

另外,美國學者研究發現,減肥也有週期,不管你有沒有刻意減肥,幾乎所有餐與實驗者,都在星期二到星期五晚上,會減少體重,到了週末,體重會自然增加,所以,建議想減肥的人,在週間嚴格執行減肥計畫,效果會相對顯著。

 

Charlie 發表在 痞客邦 留言(0) 人氣()

968218.fig.001bonekey20110507-f1EpigeneticsMechanismGene_therapy

genes.figimagesnrneurol.2013.56-f2PRFP2X7R cartoon - inline_0

痛覺由基因(Pain Genes)控制疼痛感人人不同

作者:【記者陳敬哲/綜和外電報導】 | 台灣新生報 – 201426

 

許多人有慢性或急性疼痛的困擾,長期服用止痛藥,除有成癮風險外,療效也會隨時間降低,英國倫敦國王學院研究發現,痛覺是由基因(Pain Genes)控制,每一個人疼痛感受因此有差異,若將來能針對基因開發止痛藥物,便能大幅降低疼痛困擾,慢性疼痛患者將會有更好的改善方式。

研究團隊找了二十五對同卵雙胞胎,因他們基因組成完全相同,痛覺感知不會有差異,如果出現不同,一定是受環境影響,讓基因出現變化;在實驗中,研究人員使用一個發熱的棒子,貼緊受測者的皮膚,同時提醒他們,當熱感轉為疼痛到無法忍耐,就可以按下手上按鈕,以便記錄對痛覺的耐受性。

接著研究者採取受測者的基因檢體分析,結果發現人類的基因中,有九個與疼痛相關,二十五對受測者的基因,有一對姊妹的疼痛基因有些微差距,推斷是受到環境影響,讓兩者對疼痛感知不同;這項基因變化關鍵,許多研發團隊正在開發針對性止痛藥物,效果都非常良好。

研究學者提姆教授認為,如果將痛覺比喻成電燈,基因就像調整開關,當疼痛訊號來到,必須先通過基因調控,大腦才會反應出強弱不同的痛覺,從實驗中得知,疼痛基因會隨著環境改變,未來可對此研發止痛藥物,徹底改變人體對疼痛的敏感度。

 

Charlie 發表在 痞客邦 留言(0) 人氣()

2014012900007_1Lv.1_up36989002

defaultfig2fig5fig6imagesLv.1_up36989o0618034012829169291STAP-cell-developed-01images2

日本理化學研究所發生暨再生科學綜合研究中心創舉研發出新型萬能細胞:STAP細胞

中央社 – 2014129下午10:10

(中央社記者楊明珠東京29日專電)

日本「朝日新聞」報導,理化學研究所研究人員製出新型的「萬能細胞」,研究人員將實驗鼠細胞以弱酸性液體刺激,產生初期化,變成萬能細胞。這項研究成果具劃時代意義。

包括美國哈佛大學等美日專家組成的國際研究團隊理化研等研究小組,成功地從實驗鼠體細胞製作出可變成任何一種細胞的萬能細胞。通常一旦角色確定的體細胞,接受某種程度刺激的話,不可能產生初期化,這項新的研究成果顛覆了生命科學的常識。研究成果登載於英國科學雜誌「自然」電子版。

理研的發生暨再生科學綜合研究中心(位於神戶市)的研究團隊召集人、30歲的小保方晴子等人將這種新製作成的萬能細胞稱為STAP細胞。這比誘導性多功能幹細胞(iPS細胞)能更有效率地製作,與胚胎幹細胞(ES細胞)大致一樣,不易傷害基因,所以癌化的危險性較低

京都大學鑽研萬能細胞的教授中逵憲夫對媒體表示,做為基礎研究,這研究成果令人驚訝,也讓人感興趣。這讓他再度體會到,體細胞初期化的方法還有很多深奧處,會有很多的新發現。

得過諾貝爾獎的京都大學iPS細胞研究所所長山中伸彌發表評語表示,這是一項重要的研究成果,是日本人研究有成,他引以為榮。期待今後也能從人類的細胞以相同的手法製作萬能細胞。

 

Charlie 發表在 痞客邦 留言(0) 人氣()

WEB_cover-art-suggestion-3nature12968-f1nature12968-sf7  

好簡單細胞泡澡就變幹細胞 Acid bath offers easy path to Stem cells

Stimulus-triggered acquisition of pluripotencySTAP):Just squeezing or bathing cells in acidic conditions can readily reprogram them into an embryonic state.

中央社 – 2014130

(中央社倫敦29日綜合外電報導)

美國與日本科學家今天分別在「自然」雜誌(Nature)發表研究指出,只要讓成熟動物細胞「泡澡」,就能讓細胞重新編程(reprogram)回復到幹細胞狀態並長成各種細胞組織

他們發現,如果讓成熟動物細胞處於「壓力」狀態下,它們就能回到胚胎細胞的狀態

科學家利用血液與皮膚細胞進行繁殖,並讓它們處於壓力下,「幾近死亡的臨界點」,這些壓力包括創傷、低氧與酸性環境

最簡單的方式是將細胞泡在微酸性溶液30分鐘,就能啟動回復到幹細胞狀態的過程

數天內,這些細胞不但活下來,還自然回到類似胚胎幹細胞的狀態

這些細胞之後能依所處環境,分別演變成不同細胞與組織

如果這個方法在人體上也行得通,修補受損細胞或培養新器官將更簡單。

(譯者:中央社何世煌)

 

Acid bath offers easy path to stem cells

Just squeezing or bathing cells in acidic conditions can readily reprogram them into an embryonic state.

David Cyranoski

29 January 2014

http://www.nature.com/news/acid-bath-offers-easy-path-to-stem-cells-1.14600

A mouse embryo injected with cells made pluripotent through stress, tagged with a fluorescent protein.

 

In 2006, Japanese researchers reported1 a technique for creating cells that have the embryonic ability to turn into almost any cell type in the mammalian body — the now-famous induced pluripotent stem (iPS) cells. In papers published this week in Nature2, 3, another Japanese team says that it has come up with a surprisingly simple method — exposure to Stress, including a Low pH — that can make cells that are even more malleable than iPS cells, and do it faster and more efficiently.

“It’s amazing. I would have never thought external stress could have this effect,” says Yoshiki Sasai, a stem-cell researcher at the RIKEN Center for Developmental Biology in Kobe, Japan, and a co-author of the latest studies. It took Haruko Obokata, a young stem-cell biologist at the same centre, five years to develop the method and persuade Sasai and others that it works. “Everyone said it was an artefact — there were some really hard days,” says Obokata.

Obokata says that the idea that stressing cells might make them pluripotent came to her when she was culturing cells and noticed that some, after being squeezed through a capillary tube, would shrink to a size similar to that of stem cells. She decided to try applying different kinds of stress, including heat, starvation and a high-calcium environment. Three stressors — a Bacterial toxin that perforates the cell membrane, Exposure to low pH and Physical squeezing — were each able to coax the cells to show markers of pluripotency.

But to earn the name pluripotent, the cells had to show that they could turn into all cell types — demonstrated by injecting fluorescently tagged cells into a mouse embryo. If the introduced cells are pluripotent, the glowing cells show up in every tissue of the resultant mouse. This test proved tricky and required a change in strategy. Hundreds of mice made with help from mouse-cloning pioneer Teruhiko Wakayama at the University of Yamanashi, Japan, were only faintly fluorescent. Wakayama, who had initially thought that the project would probably be a “huge effort in vain”, suggested stressing fully differentiated cells from newborn mice instead of those from adult mice. This worked to produce a fully green mouse embryo.

Still, the whole idea was radical, and Obokata’s hope that glowing mice would be enough to win acceptance was optimistic. Her manuscript was rejected multiple times, she says.

To convince sceptics, Obokata had to prove that the pluripotent cells were converted mature cells and not pre-existing pluripotent cells. So she made pluripotent cells by stressing T cells, a type of white blood cell whose maturity is clear from a rearrangement that its genes undergo during development. She also caught the conversion of T cells to pluripotent cells on video. Obokata called the phenomenon Stimulus-triggered acquisition of pluripotency (STAP).

The results could fuel a long-running debate. For years, various groups of scientists have reported finding Pluripotent cells in the mammalian body, such as the Multipotent adult progenitor cells described4 by Catherine Verfaillie, a molecular biologist then at the University of Minnesota in Minneapolis. But others have had difficulty reproducing such findings. Obokata started the current project in the laboratory of tissue engineer Charles Vacanti at Harvard University in Cambridge, Massachusetts, by looking at cells that Vacanti’s group thought to be pluripotent cells isolated from the body5. But her results suggested a different explanation : that Pluripotent cells are created when the body’s cells endure physical stress. “The generation of these cells is essentially Mother Nature’s way of responding to injury,” says Vacanti, a co-author of the latest papers2, 3.

One of the most surprising findings is that the STAP cells can also form Placental tissue, something that neither iPS cells nor embryonic stem cells can do. That could make cloning dramatically easier, says Wakayama. Currently, cloning requires extraction of unfertilized eggs, transfer of a donor nucleus into the egg, in vitro cultivation of an embryo and then transfer of the embryo to a surrogate. If STAP cells can create their own placenta, they could be transferred directly to the surrogate. Wakayama is cautious, however, saying that the idea is currently at “dream stage”.

Obokata has already reprogrammed a dozen cell types, including those from the brain, skin, lung and liver, hinting that the method will work with most, if not all, cell types. On average, she says, 25% of the cells Survive the stress and 30% of those convert to Pluripotent cells — already a higher proportion than the roughly 1% conversion rate of iPS cells, which take several weeks to become pluripotent. She now wants to use these results to examine how reprogramming in the body is related to the activity of stem cells. Obokata is also trying to make the method work with cells from adult mice and humans.

“The findings are important to understand nuclear reprogramming,” says Shinya Yamanaka, who pioneered iPS cell research. “From a practical point of view toward clinical applications, I see this as a new approach to generate iPS-like cells.”

 

Nature 505, 596 (30 January 2014) doi:10.1038/505596a

 

References

Takahashi, K. & Yamanaka, S. Cell 126, 663–676 (2006).

Obokata, H. et al. Nature 505, 641–647 (2014).

Obokata, H. et al. Nature 505, 676–680 (2014).

Jiang, Y. et al. Nature 418, 41–49 (2002).

Obokata, H. et al. Tissue Eng. Part A 17, 607–615 (2011).

Charlie 發表在 痞客邦 留言(0) 人氣()

Autotransfusion with RBC and Platelet-Rich PlasmaIntraoperative Autotransfusion2

3. QUALITY OF CELLS, ADVERSE EFFECTS

3.1. WASHED BLOOD

Cell separators remove Plasma from salvaged shed blood, eliminating potentially Toxic products of injured red cells and procoagulants <|[21]|>. Thus, the RBC concentrate is virtually Devoid of any particulate matter, including plasma proteins, coagulation factors and platelets (except for a small, clinically non-relevant fraction of nonfunctional cells and residual heparine) <|[10, 21]|>. Leucocytes are discharged to a varying degree, depending on the IAT device used <|[22]|>. Most of the Endogenous compounds released at the operative site and substances introduced by the surgeon are Washed out, as well as anticoagulants, antibiotics and other systemically administered drugs <|[23-25]|>. Conversely, some other endogenous Byproducts may be Concentrated (catecholamines, anaphylatoxines, leukotrienes, endotoxines) <|[25, 26]|>. Especially in orthopedic cases, wound blood frequently contains nonemulsified fat from bone marrow or subcutaneous depots which may ultimately cause venous fat embolism <|[11]|>. Devices based on discontinuous centrifugation cannot remove or filter out fat particles. Continuously processing cell separators are more effective in clearing fat and other contaminants such as inflammatory mediators <|[11, 14, 15]|>.

Excessive retransfusion of salvaged RBC and Saline may cause significant hematologic changes : Electrolyte and Acid-base balance may be disturbed by increase of sodium and chloride serum levels, and decrease of magnesium, ionized calcium, pH, pCO2, protein and albumine <|[25]|>. Extended IAT use may also cause a certain degree of hemolysis. While the consecutively augmented potassium release is counteracted by the washing process <|[11]|>, no cell separator can provide a complete clearence of Free hemoglobin, and serum levels may temporarily be elevated <|[8, 14, 15, 27]|>. Furthermore, since washed blood does not contain coagulation factors and platelets, massive RBC autotransfusion results in Dilutional coagulopathy and a Decrease of circulating platelets, requiring administration of the appropriate allogeneic components <|[6, 8, 19, 28]|>. Coagulation may further be disturbed by Prolonged hypothermia and shock, as well as by Reinfusion of residual heparin <|[1, 28]|>. Other adverse effects, such as Air embolism, Microembolism and Changes in 2,3-DPG affecting oxygen-carrying capacity of the red cells <|[8]|>, have become extremely rare if nonexistent after the arrival of electronically controlled, up-to-date cell separation devices.

 

3.2. UNWASHED BLOOD

Retransfusion of Unwashed autologous whole blood has been demonstrated to be safe and equieffective to Cell separation <|[16-19, 21]|>. However, the Quality of unprocessed blood is Inferior with a Higher return of lysed cellular debris, platelet microaggregates, fibrin split products, d-dimer, activated complement and free hemoglobin, indicating imbalanced coagulation due to pronounced hemostatic activation and increased hemolysis <|[Table I]|> <|[6, 19, 21, 27]|>. Especially in patients systemically anticoagulated for surgical purposes, simple IAT may not be detrimental in smaller amounts <|[6,19]|>, but larger quantities may lead to severe hemostatic disturbances with the potential of triggering Disseminated intravascular coagulation <|[6]|>. Because of this limited tolerance to unwashed wound blood, the total amount of retransfusion should clearly be restricted <|[6, 18, 21, 27]|>.

 

4. INDICATIONS

Intraoperative cell salvaging should be considered for all acute Emergency procedures. In severely traumatized patients with intense hemorrhage, the use of IAT may literally be life-saving due to a possibly limited supply of allogeneic blood to compensate for extreme blood loss. For practical purposes, the proper handling and processing of allogeneic blood transfusions in severe emergency situations may also pose substantial logistic challenges, thus supplemental autologous RBC supply provided by the comparatively simple IAT procedure may be extremely helpful. For these reasons, it is recommended that at least one IAT device should always be available and prepared in emergency rooms and operating units, and the attending staff must be well-trained in its proper use <|[4]|>.

In elective surgery, the routine application of IAT is controversial because of unpredictable cost-effectiveness <|[29, 30]|>. IAT has been shown to work efficiently in major orthopedic <|[9, 12, 31]|>, cardiac <|[32]|>, urologic <|[33, 34]|>, vascular <|[27]|>, intracranial <|[35]|> and gynecological <|[36]|> surgery and in emergency procedures, such as spinal trauma <|[37]|>. However, due to the wide variety of surgical, patient-related and institutional factors, it is difficult to define specific surgical procedures where IAT should always be used, and the overall blood salvage may not cover more than 25% of all perioperative blood losses <|[5, 12]|>.

<|[Table II]|> lists major surgical specialties where IAT can be successfully used and may be implemented in the perioperative blood saving strategies. As one practicable clinical guideline, the use of IAT is recommended for all major surgical procedures with anticipated blood loss of ≥ 1,000 ml <|[2, 5, 29]|>. At this level, IAT becomes cost-effective, but patients who are estimated to lose less than 1,000 ml are likely to receive little benefit yet incur substantial costs <|[29]|>. On the other hand, cost-effectiveness calculations limited to short-term perspectives do not take into account the enormous additional costs that may arise due to transmission of viral diseases or other adverse effects related to allogeneic transfusions <|[5, 38]|>.

Unfortunately, estimates of allogeneic blood requirement by preoperatively available data are very difficult and unreliable, even for planned, uncomplicated surgery <|[12, 39]|>. The anesthesiologists individual experience and knowledge of institutional circumstances (based on retrospective analyses), the type of procedure, and the patients red cell reserve, body mass and gender are yet the most helpful parameters in predicting intraoperative blood loss <|[12]|>. One rather elegant solution to this dilemma is the use of inexpensive intermediate storage bags whenever blood loss cannot be properly anticipated. Anticoagulated shed blood is put on hold, and when the amount of collected blood is sufficient to justify the use of IAT, it can be transferred to the cardiotomy reservoir and be further processed <|[4]|>. This strategy allows to save costs, while providing maximal patient safety.

Charlie 發表在 痞客邦 留言(0) 人氣()

550px-AUTOTRANSFUSIONPD

Fresenius CATS Diagram copy  

Autotransfusion with RBC and Platelet-Rich PlasmaIntraoperative Autotransfusion1

http://www.nataonline.com/node/433

 

Abbreviations

CPBcardiopulmonary bypass

PRPplatelets poor plasma

IATintraoperative autotransfusion

IPintraoperative plasmapheresis

RBCred blood cells

 

A. RBC

1. BACKGROUND SUMMARY

Intraoperative autotransfusion (IAT) is defined as the reinfusion of patient blood salvaged during and after surgery. IAT plays an important role in the context of perioperative blood saving strategies, having assumed standard of care status for many surgical procedures <|[1-6]|>.

The principle of IAT is to continuously collect intra- or post-operatively shed blood from the operative field. The salvaged blood is aspirated from the wound site and collected in a dedicated reservoir. Under standard conditions, Red cells are subsequently Separated, Washed, Hemoconcentrated and Stored for subsequent retransfusion to the patient. Only Erythrocytes are saved and retransfused, thus simultaneous volume and plasma replacement has to be provided, especially after processing of large quantities of shed blood <|[4]|>.

In contrast to stored RBC, freshly salvaged autologous red cells show Uncompromized functional capacity, oxygen delivery to tissues and survival, indicating that IAT has no significant detrimental effects on erythrocytes <|[7, 8]|>. IAT is most effective when combined with other autologous methods, particularly with Pre-operative autologous blood donation, Acute normovolemic hemodilution or Adiuvant drug therapies <|[4, 9, 10]|>.

 

2. METHODS

2.1. CELL SEPARATION (WASHED BLOOD)

This technique is based on Centrifugation, Separating red blood cells (RBC) from the Lighter components and fluids, including plasma, saline and buffy coat (Cell saver, Haemolite, Haemonetics, Braintree, MA; <|[figure 1]|>. Before starting the procedure, the system has to be filled with 100200 ml Heparinized saline (Priming), in order to Prevent cells from binding to membrane surfaces initiating Microaggregation, and to Diminish frictional forces and Damage to the cellular components <|[4, 8]|>. Blood released at the wound site is aspirated via a double-lumen suction catheter (80-100 mmHg), immediately anticoagulated (30,000 IU Heparin in 1,000 ml Saline) at the suction tip, and stored in a plastic cardiotomy reservoir, equipped with a microaggregate 120 m filter. When a minimum of 1,000 ml shed blood is collected, it is pumped into a rotating separation chamber (Latham bowl, 225 ml adaptable capacity), Washed with 10001500 ml Saline and concentrated. Whenever the extent or kind of surgical debris requires more extensive washing, processing cycles can be selected manually, and for emergency cases, washing can be skipped entirely. For pediatric patients, smaller centrifuges are available. As soon as the preset hematocrit is reached, the spinning separator chamber stops, and packed RBC, suspended in Saline solution, are pumped into an infusion bag, while the waste products are removed <|[4, 11]|>. After completion of each cycle, the bowl can be filled again for as many times as required. The Hematocrit of the final erythrocyte supension is regulated by an optical sensor in the centrifugation chamber, targeting 5570% of packed cell volume <|[4, 5, 11-13]|>.

Modified cell separation can also be performed by passing collected blood repeatedly through a vortex mixing filter with longitudinal channels and microporous plastic membranes (Haemocell System 350, Haemonetics), under continuous washing with saline solution <|[8]|>. Apparently, this less traumatizing method seems to have minor detrimental effects on the shed blood, better preserving the function of red cells and platelets <|[8]|>. However, no comparative studies have been performed up to now.

A recently developed novel device (Continuous AutoTransfusion System; Fresenius, Bad Homburg, Germany; <|[figure 2]|> allows continuous blood processing instead of operating in batches or units <|[11, 14]|>. The separation chamber used in this model represents a blood channel in the shape of a double spiral (capacity approx. 30 ml). Blood is pumped into the inner loop, while the separation chamber rotates continuously. Substances of less density leave the spiral immediately at this point, while RBC are moved towards the outer spiral, being continuously washed with saline <|[11, 14, 15]|>. All steps are performed simultaneously, allowing immediate retransfusion, even of smaller amounts, of processed red cells. Because of the maintained rotation of the separation unit, RBC cannot remix with lighter particles (eg. fat2). Thus, this novel cell separator may be more efficient in clearing substances that have been difficult to remove with conventional methods <|[11, 14]|>.

Disadvantages of cell separators might be less obvious : time and human resources are required for the setup, and the disposable single-use sets are expensive. For cases when blood loss may not be sufficient to warrant IAT operation, simple and inexpensive collection bags are available, allowing temporary storage of shed blood until further processing.

 

2.2. SIMPLE AUTOTRANSFUSION (UNWASHED BLOOD)

Retransfusion of Unwashed, Filtered whole blood (Srensen, Solcotrans, Biosurge) has been a clinically acclaimed alternative, both in regard to costs, ease of use and effectiveness in returning shed wound blood <|[16-19]|>. However, these devices have primarily been designed for the salvage of slowly oozing blood rather than rapid hemorrhage <|[8]|>. With the evolvement of new generations of safe, fully automated and easy-to-use cell separators, the importance of simple autotransfusion has largely shifted towards the postoperative phase <|[4, 6, 20]|>.

<|[Table I]|> summarizes typical properties of simple autotransfusion vs. cell separation devices <|[4]|>.

Charlie 發表在 痞客邦 留言(0) 人氣()

300px-Eicosanoid_synthesis.svgimage002Layout-4_Pagina-124nature12085-f1.2nri2395-f3scrt2-1scrt64-1-l

調經用藥PGE2 讓老化心肌再生

作者:洪榮志台南報導 | 中時電子報 – 2014128

中國時報【洪榮志台南報導】

 

醫學界普遍認為心肌無法再生,但成功大學團隊27日指出,歷經7年的研究發現,以婦產科調整月經周期的前列腺素E2(簡稱PGE2)治療心臟衰竭時,不僅可增進年輕個體心臟幹細胞的修復效率,還能恢復老化個體心肌的再生能力,徹底顛覆醫學界過去的認知。

此項研究係由成大臨床醫學所教授謝清河帶領博士生薛盈彰和吳美芳共同研究,除已刊登於歐洲分子醫學期刊(EMBO Molecular Medicine)外,還申請多國專利,正準備進一步開發抗老化、促進心肌再生的新藥。

謝清河表示,心臟疾病高居全球死亡率排名第1位,在台灣也是國人10大死因第2位,僅次於癌症。目前面對心臟衰竭的病人,無論採用內科或外科的治療方式,效果均十分有限;尤其是對於末期心臟衰竭的病人,只剩換心一途才能挽救生命。

該團隊發現,年輕老鼠在心肌梗塞發生後3天內,就會啟動自體幹細胞心肌修復的機制;在梗塞後710天達到高峰,之後幹細胞修復的機制就停止進行。

該團隊從早期發炎反應抽絲剝繭,發現PGE2就是發炎反應啟動幹細胞再生的關鍵因子,缺乏PGE2訊息傳遞路徑,幹細胞就會喪失再生能力。相反地,於心臟受損後給予PGE2治療,則可增強原有心肌再生的效率

有趣的是,該團隊以基因轉殖鼠實驗,發現年老老鼠的心臟缺乏幹細胞再生心肌能力,並非年老個體缺乏幹細胞、或幹細胞數量減少的緣故,而是幹細胞本身的活性被周圍老化的微環境所抑制,以致無法發揮作用

一旦PGE2移除掉這些老化的因子,就能讓幹細胞重獲生機,返老還童,恢復心肌再生的能力。

 

Charlie 發表在 痞客邦 留言(0) 人氣()

autismAUTISM_brainInfographic2Autistic-Males-Lack-Neurons-in-the-Amygdala-2image_mediumpicture-101

中研院揭秘自閉症肇因杏仁核神經迴路異常

自由時報 – 2014128

〔自由時報記者湯佳玲/台北報導〕

自閉症治療出現新契機!中研院分子生物研究所研究員薛一蘋的研究團隊,首度發現自閉症致病機制,一旦主管社交活動的腦中杏仁核神經迴路缺少「後段前連合」,就會出現自閉症狀,現有肺結核治療藥物「D-環絲胺酸」可以有效治療類自閉症小鼠的異常行為,為自閉症治療開啟一扇窗。

 

國內自閉症人數逾二萬二千人

內政部統計,國內2012年自閉症人數已達二萬二千多人,較十年前成長約2.7倍,是各類身心障礙者中人數增幅較大的。美國每八十八位兒童即有一位自閉兒,男童發生率更為女童五倍左右。

薛一蘋表示,臨床上已知自閉症與神經迴路有極大關係,也已知控制大腦皮質及杏仁核發育的TBR1基因與自閉症有關,但並不了解TBR1突變如何引發自閉症。

 

「後段前連合」缺陷致病主因

她的團隊選用「TBR1缺失」的小鼠來模擬人類缺少TBR1的生理變化,發現這些小鼠會因此喪失腦中特定的「後段前連合」蛋白質結構,導致大腦中掌控社交活動、情緒反應、恐懼記憶等功能的杏仁核無法彼此聯繫,造成神經迴路異常,訊息無法正確傳遞。顯示一旦「後段前連合」出現缺陷,就會影響杏仁核神經迴路,表現出類似自閉症的行為特徵。

研究團隊在小鼠腦部的杏仁核直接注射D-環絲胺酸藥物,去活化TBR1所控制的離子通道活性,發現原本失去社交活動、不太理會同籠其他同伴的基因缺失鼠,又會追著其他小鼠的尾巴跑了。

 

肺結核藥可治療仍須臨床試驗

薛一蘋說,D-環絲胺酸是現有治療肺結核的抗生素藥物,市面上已使用五十年,且通過人體試驗,但「舊藥新用」發現對於治療自閉症也有效。「它雖無法讓『後段前連合』長回來,但會刺激離子通道活化,讓活性變得更多、更好,控制成正常的神經表現。」目前D-環絲胺酸用在小鼠身上有效,但仍需經過臨床試驗。該藥對人體無害,現為醫師開立的處方用藥,相信很快就能成為人體自閉症的臨床用藥。

薛一蘋認為,未來病人只需做非侵入性MRI核磁共振造影,看看是否「後段前連合」有缺陷,就能與醫師搭配使用D-環絲胺酸,治療自閉症。

 

Charlie 發表在 痞客邦 留言(0) 人氣()

floseal-ifu-comparisonSurgiflo_SNGtisseelUSP1011-Hemostat-F1  

止血凝膠(Hemostatic Matrix)提升手術視野人工膝關節置換免驚

作者:台北訊 | 中時電子報 – 2014127

中國時報【台北訊】

(中時健康趙欣報導)

台灣老年人口比例日益增加,根據行政院主計總處的統計,截至民國103年底,台灣65歲以上的老年人口總計260餘萬人,且正在快速成長中。前陣子於立法院通過初審的長期照護服務法草案,更是因應台灣人口快速老化的政策之一。

觀察老年人的健康狀況,關節正是問題的大宗,特別是退化性關節炎造成了許多年長者生活上極大的不便。除了上下樓梯困難,嚴重者甚至連日常行走都會感到疼痛難耐。日前一名年約60歲的蔡先生,因為工作的需要經常往返國內外,但卻因膝蓋問題影響行動,因此在最近半年內接受了兩次人工膝關節置換手術。

第一次手術時,為了日後行走方便也考量到適應問題,蔡先生僅於右腳接受了微創手術加上引流管,住院一週以後便回家修養。第二次於左腳手術時,他聽取了醫師的建議,搭配使用止血凝膠,約4天左右便出院,且腫脹程度較小,令他十分滿意。

骨科醫師江福財指出,除了自身老化的因素,體重是造成膝關節磨損最主要的原因,而肥胖導致膝關節退化的速度,比其他原因都要來得更快。目前接受人工膝關節置換手術的國人,以70歲左右為大宗,另外有近四分之一是介於5060歲間的中老年人。

針對置換人工關節的評估,骨科醫師江福財表示,一般會先透過X光片來診斷病人的情況,若是膝關節的三個關節面當中,已有兩個損壞嚴重,就會建議更換,若僅有一個損傷,則會建議追蹤觀察即可。不過,由於人工膝關節置換是屬創傷面比較多的一種手術,過程中出血量較大,要是沒有做好止血工作,血液堆積在膝關節腔中,就可能會導致腫脹或血腫的情形,過去甚至曾出現失血過多需要緊急輸血的個案。

對此,江福財醫師表示,目前國內膝關節置換手術的技術已十分成熟,成功率也相當高,惟須特別留意出血狀況。過往多依賴凝血藥物,但效果有限,近年則出現止血凝膠,輔助不小。止血凝膠,顧名思義是種能創造止血環境的成分,一般只要在手術中,於出血點覆蓋流體冰沙狀的凝膠,就能達到快速止血的效果。一來減低出血量,讓醫師於手術中的視野更清晰,二來則能減低後續腫脹或血腫的機會。

對於部分民眾憂心若是患有嚴重肝硬化或慢性腎衰竭,導致凝血功能不全,是否仍適用止血凝膠的問題。江福財醫師說,止血凝膠對於凝血功能不全或年紀較大的患者來說都是適用的,一般手術後68週就會被人體自然吸收,且目前臨床上沒有產生副作用的紀錄。值得注意的是,由於內含明膠以及凝血酶(Thrombin)等成份,其中,明膠是來自於牛皮的組織粹取(Gelatin Matrix),被廣泛應用於藥品膠囊的製造,對其過敏的機率極低,但若是對牛蛋白過敏者就無法使用。

Charlie 發表在 痞客邦 留言(0) 人氣()

IMG_2133image002image004image007m04

Placental_cord_cross-sectionRE054bstemcell_umbilical_09

umbilicalcord-crosssection3

troyer_image

Wharton's jelly

Wharton's jellysubstantia gelatinea funiculi umbilicalisis a gelatinous substance within the Umbilical cord, also present in Vitreous humor of the eyeball, largely made up of mucopolysaccharidesHyaluronic acid and Chondroitin sulfate.

It also contains some fibroblasts and macrophages.

It is derived from extra-embryonic mesoderm.

As a mucous tissue it protects and insulates umbilical blood vessels.

Wharton's jelly, when exposed to temperature changes, collapses structures within the umbilical cord and thus provides a physiological clamping of the cord an average of 5 minutes after birth.

Cells in Wharton's jelly express several stem cell genes, including telomerase. They can be extracted, cultured, and induced to differentiate into mature cell types such as neurons.

Wharton's jelly is a potential source of adult stem cells.

Wharton's jelly is named for the English physician and anatomist Thomas Wharton (1614–1673) who first described it in his publication Adenographia, or "The Description of the Glands of the Entire Body", first published in 1656.

 

Charlie 發表在 痞客邦 留言(0) 人氣()

0T5323003-01_101230095243_11-12020611414W401504px-Missingself.svg1390217051-134036902_n1390217051-1699325614_n1390217051-3861207462_n1390217051-3867178808_n1390217051-40697962862010471650422760

default

CIK cellCytokine-induced Killer cells

A preparation of autologous Lymphocytes with potential immunopotentiating and antineoplastic activities.

Cytokine-induced killerCIKcells are CD3-and CD56-positive, non-Major histocompatibility complexMHC-restricted, Natural killerNK-like T lymphocytes, generated ex-vivo by incubation of peripheral blood lymphocytesPBLswith anti-CD3 monoclonal antibody, interleukin-2IL-2, IL-1, and interferon gammaIFN-gammaand then expanded.

When reintroduced back to patients after autologous stem cell transplantation, CIK cells may recognize and kill tumor cells associated with minimal residual diseaseMRD.

CIK cells may have enhanced cytotoxic activity compared to lymphokine-activated killerLAKcells.

 

Charlie 發表在 痞客邦 留言(0) 人氣()

2157-7633-1-105-g0012157-7633-2-e107-g001200510en_4ar2103-1bonfield_caplan_no47_figure_1enmesenchymalstem1  

MSCMesenchymal Stem Cell or Multipotent Stromal Cell

Mesenchymal stem cells, or MSCs, are multipotent stromal cells that can differentiate into a variety of cell types, includingOsteoblasts (bone cells), Chondrocytes (cartilage cells), and Adipocytes (fat cells). This phenomenon has been documented in specific cells and tissues in living animals and their counterparts growing in tissue culture.

While the terms Mesenchymal Stem Cell and Marrow Stromal Cell have been used interchangeably, neither term is sufficiently descriptive as discussed below

Mesenchyme is embryonic connective tissue that is derived from the mesoderm and that differentiates into hematopoietic and connective tissue, whereas MSCs do not differentiate into hematopoietic cells.

Stromal cells are connective tissue cells that form the supportive structure in which the functional cells of the tissue reside. While this is an accurate description for one function of MSCs, the term fails to convey the relatively recently discovered roles of MSCs in the repair of tissue.

Because the cells, called MSCs by many labs today, can encompass multipotent cells derived from other non-marrow tissues, such as umbilical cord blood, adipose tissue, adult muscle, corneal stroma or the dental pulp of deciduous baby teeth, yet do not have the capacity to reconstitute an entire organ, the term Multipotent Stromal Cell has been proposed as a better replacement.

The youngest, most primitive MSCs can be obtained from the Umbilical cord tissue, namely Wharton's jelly and the Umbilical cord blood. However the MSCs are found in much higher concentration in the Wharton’s jelly compared to the Umbilical cord blood, which is a rich source of Hematopoietic stem cells. The umbilical cord is easily obtained after the birth of the newborn, is normally thrown away and poses no risk for collection. The umbilical cord MSCs have more primitive properties than other adult MSCs obtained later in life, which might make them a useful source of MSCs for clinical applications.

An extremely rich source for mesenchymal stem cells is the developing Tooth bud of the Mandibular third molar. While considered multipotent, they may prove to be Pluripotent. The stem cells eventually form enamel, dentin, blood vessels, dental pulp, nervous tissues, including a minimum of 29 different unique end organs. Because of extreme ease in collection at 8–10 years of age before calcification and minimal to no morbidity they will probably constitute a major source for personal banking, research and multiple therapies. These stem cells have been shown capable of producing Hepatocytes. Additionally, Amniotic fluid has been shown to be a very rich source of stem cells. As many as 1 in 100 cells collected from and genetic amniocentesis has been shown to be a Pluripotent mesenchymal stem cell.

Adipose tissue is one of the richest sources of MSCs. When compared to bone marrow, there is more than 500 times more stem cells in 1 gram of fat when compared to 1 gram of aspirated bone marrow. Adipose stem cells are currently actively being researched in clinical trials for treatment in a variety of diseases.

The presence of MSCs in peripheral blood has been controversial. However, a few groups have successfully isolated MSCs from human peripheral blood and been able to expand them in culture.

In 1924, Russian-born morphologist Alexander A. Maximow used extensive histological findings to identify a singular type of precursor cell within mesenchyme that develops into different types of blood cells.

Scientists Ernest A. McCulloch and James E. Till first revealed the clonal nature of marrow cells in the 1960s. An ex vivo assay for examining the clonogenic potential of multipotent marrow cells was later reported in the 1970s by Friedenstein and colleagues. In this assay system, stromal cells were referred to as colony-forming unit-fibroblasts (CFU-f).

Subsequent experimentation revealed the plasticity of marrow cells and how their fate could be determined by environmental cues. Culturing marrow stromal cells in the presence of osteogenic stimuli such as ascorbic acid, inorganic phosphate, and dexamethasone could promote their differentiation into Osteoblasts. In contrast, the addition of Transforming growth factor-beta (TGF-b) could induce Chondrogenic markers.

The mesenchymal stem cells can be activated and mobilized if needed. However, the efficiency is very low. For instance, damage to muscles heals very slowly. However, if there were a method of activating the mesenchymal stem cells, then such wounds would heal much faster.

Many of the early clinical successes using intravenous transplantation have come in systemic diseases like graft versus host disease and sepsis. However, it is becoming more accepted that diseases involving peripheral tissues, such as inflammatory bowel disease, may be better treated with methods that increase the local concentration of cells. Direct injection or placement of cells into a site in need of repair may be the preferred method of treatment, as vascular delivery suffers from a "Pulmonary first pass effect" where intravenous injected cells are Sequestered in the lungs. Clinical case reports in orthopedic applications have been published, though the number of patients treated is small and these methods still lack rigorous study demonstrating effectiveness. Wakitani has published a small case series of nine defects in five knees involving surgical transplantation of mesenchymal stem cells with coverage of the treated chondral defects.

Charlie 發表在 痞客邦 留言(0) 人氣()