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()

300px-Location_of_Buccal_Fat_Pad1378798808-20614681601378798815-1730946724Buccal_Fat_DiagramF3.largeJNatScBiolMed_2012_3_2_203_101930_f3  

天生嬰兒肥?「口內取脂」成功瘦小臉!

【中時健康任呈岳/台北報導】2014.02.21

每個人對可愛的認知不同,但是即便有一張BABY FACE的外表,還是希望能有美女般的瓜子臉型!醫師提醒,造成嬰兒肥的原因很多,若想改善,還是要找專業醫師進行研判,並依照每個人不同臉型進行整形評估,現在也能從口中進行抽脂,傷口小、恢復時間短,對外觀也沒有影響,也能列為選擇之一。

Vickey天生就有一張娃娃臉,年輕時,兩頰肉肉的嬰兒肥,更讓她成為男生都想保護的小妹妹。但年過三十歲,BABY FACE反倒成為困擾。無論怎麼減重與利用小臉器,就是無法讓臉頰的兩塊肉消失,直到醫師告訴她原因其實是出在於兩頰深層的脂肪墊肥厚,才讓她恍然大悟。

葛萊美診所整形醫美中心院長吳名倫醫師指出,雖然嬰兒肥看來可愛,不過對於追求瓜子臉的民眾而言,可能是一種夢靨,然而,許多人卻對於嬰兒肥有錯誤觀念,其中最多人犯的錯誤,即是以為「嬰兒肥就是咀嚼肌太過發達」,結果經常施打肉毒桿菌,治療成果當然不盡理想

造成嬰兒肥的原因,其實大多是出在兩頰深層的脂肪墊太過肥厚所致,民眾可自我分辨到底是脂肪墊肥厚還是咀嚼肌過於發達。吳名倫醫師建議,大家可以照鏡子從正面觀察,若為兩頰脂肪墊肥厚所造成的,膨起位置會在臉部上緣且比較前面;至於若是咀嚼肌肥大,看起來則是膨起位置會比較後面、且在臉部下緣的兩側。

目前針對圓臉的治療方式有很多種,包括肉毒桿菌、削骨手術、齒顎矯正等,但若是單純的雙頰深層脂肪墊肥厚,就可以採用傷害性較小的「口內取脂」手術。吳名倫醫師解釋,所謂口內取脂,就是從口腔內的雙頰,各開一個一公分不到的傷口,將深層的脂肪墊取出。

比起一般的抽脂手術,醫師在施行口內脂肪墊取出手術時,是直接由口內的傷口內目視取出脂肪墊,不會傷及神經、大血管等組織,更不會在臉部外觀上留下任何傷口或疤痕,是一個風險度很低的手術;但由於口內還是會有傷口,因此,術後可能要比照牙齒手術,對於傷口的清潔消毒非常重要,術後一周內也不能食用刺激性食物,進食後要多漱口以保持口腔的清潔。

當然,把口內脂肪墊取出並不代表可解決所有的圓臉問題,書田診所皮膚科主任鄭惠文醫師提醒,臉部的整型必須用一個整體角度予以判斷,不可能單靠一個手術就能解決所有問題,最好選擇有較多經驗的醫師,依照每個人不同的條件與需求,進行整體的調整計劃,才能讓治療事半功倍!

Charlie 發表在 痞客邦 留言(0) 人氣()

2014223L09M1  

140221-2  

保養品號稱「修復皮膚」,甚至「肌因賦活」,或「胜抗老成分」1%滲入皮膚

民視 – 2014223

 

市面上販售保養品大多宣稱可「修復皮膚」,甚至標示「肌因賦活露」,或「胜抗老成分」,讓許多人誤以為真有功效,但皮膚科醫師說,多數宣稱可「修復」的保養品,其實滲進皮膚比率不到1%,食藥署現在打算針對市售標榜「肌因修復」等商品進行調查,考慮修法,不讓業者鑽漏洞,誤導消費者。

塗塗又抹抹,保養品擦在皮膚上,無論是雙手或臉頰,希望皮膚變好,但現在市面上有許多保養品宣稱可修復或抗老化,還有廠商將保養品翻譯成「肌因賦活露」,讓民眾真以為有這麼神奇,修復酵素在一般使用下,滲透進皮膚的比率不到1%,要是加量使用,滲透進皮膚的比率也不會超過5%,因此成分要進入基因,難度相當高,但是一般保養品如果只標示「潤膚保養」,會讓人覺得沒什麼吸引力,因此廠商才創造出「促進修復、延緩老化」等字眼,但大都是心理作用,成效不大。

醫師說臨床上具有保護成分的有,凡士林、乳油木,具修復成分的保養品有玻尿酸鹽和蘆薈萃取,食藥署強調目前市售上百種保養品,外包裝宣稱抗老、凍齡、再生、修補、重建等功能性字眼,都不允許標示,但業者巧妙地用同音或同意字代替,遊走法律邊緣,現在食藥署考慮研擬修法,不讓業者鑽漏洞,誤導消費者。

Charlie 發表在 痞客邦 留言(0) 人氣()

_

20090918074738106

美國研究:女人挑老公首要條件 品格比帥氣外型更重要

NOWnews – 2014223日國際中心/綜合報導

 

擁有一張英俊帥氣臉孔的男性,在感情關係裡或許異性緣會較他人來的好,不過,近期有項研究發現,其實女性在挑選結婚對象時,會將品格看得比外表重要。

根據英國《鏡報》(The Mirror報導,美國研究團隊以13千多名年齡低於34歲的男女為分析對象,且這些受訪者皆有超過15年以上的戀愛經驗,研究結果發現,女性在挑選男友與結婚對象時的條件並非全然相同

研究指出,女性在挑選男友或是同居對象時,長相較為英俊的男性確實可得到較多關注與偏愛,不過若是在選擇結婚伴侶時,外型帥氣的男性就不一定吃香,因為女性在挑選能夠一起跨入婚姻階段的伴侶時,會將品格良好的男性當成首選

研究人員表示,女性在挑選結婚伴侶時會多方面要求,而不只是看對方的臉蛋及外型,若是對方擁有很好的品格,儘管外型並非完美或有部分缺陷,女性也不會太在乎。

 

Charlie 發表在 痞客邦 留言(0) 人氣()

Pic_GHK_Cu_21620520_586863204716506_413790768_n  

「胜肽」、「生長因子」、「修復酵素」皮膚修復效果缺實證

作者:【中央社台北二十二日電】 | 台灣新生報 – 2014223

 

坊間促進皮膚修復保養品種類多且價格高。皮膚科醫師邱品齊說,「胜肽」、「生長因子」、「修復酵素」修復保養品效果仍未知。

台灣再生醫學學會學術研討會今天舉行,台大醫院雲林分院皮膚科主任邱品齊報告「促皮膚修復化粧品之現狀簡介」。

邱品齊說,過去化粧品強調遮蓋修飾,現今潤膚保養是基本常識,隨時代及科技,坊間出現許多促進修復產品,而臨床上相關產品不多,不過修復議題在保養品市場非常受到重視,也出現許多強調修復功能高價產品。

他說,坊間標榜含「胜肽」、「生長因子」、「修復酵素」的修復產品至今相關研究文獻不多,也都只是體外細胞研究,但沒有加入其他成分,然而保養品都是很多成分,混合一起也會讓效能減弱,也無法類推或應用在皮膚修復。

另,法規對於生長因子使用在皮膚的規範力弱,也沒有限定修復酵素的醫療用途,因此讓廠商有模糊空間,也易造成民眾困擾。

邱品齊表示,生長因子前身是胜肽,胜肽效果有限,且其結構的分子組合是不穩定,易被水解,因此難達效果;生長因子分子大、吸收不易需調控

邱品齊指出,民眾對於「陌生」、「新的」名詞或形容詞就會覺得很厲害、很神,日前購物台銷售4GF,就是把四種生長因子加在一起,但是這不是辦家家酒,每項對細胞修復是不同的。邱品齊說,雖然法規沒禁止前述產品,不過的確還有很多缺乏實證資料,提醒想要使用的民眾「不要期待過高」。

邱品齊舉例具修復功能的成分包括金屬類鹽類(鋅、銅、錳)、雷公根萃取、蘆薈萃取、甘草次酸、維他命AB3B5CE等,而且文獻討論也比較多。邱品齊指出,皮膚本身就有修復功能,因此只要做好保濕及防曬,若有傷口則給予油脂類保護,例如凡士林,只要皮膚自己保護好了,就會啟動修復功能。

Charlie 發表在 痞客邦 留言(0) 人氣()

t3_20140223.600_148  

「肌因」、「DNA修復」「促進修復」、「延緩老化」、「抗老、凍齡、再生、修補、重建」保養品修復皮膚?「別被迷惑」

自由時報 – 2014223

〔自由時報記者邱宜君/台北報導〕

「肌因」、「DNA修復」「促進修復」、「延緩老化」、「抗老、凍齡、再生、修補、重建」?保養品名詞炫別被迷惑

 

坊間很多宣稱可修復皮膚、甚至再生的保養品,價位高,成分也很神秘,但民眾無從確知是真或假?台大醫院雲林分院皮膚科主任邱品齊提醒,不要被「肌因」或「DNA修復」等名詞給迷惑了。

現代人追求速效、喜新厭舊,化妝品基本的潤膚保養功能已經沒什麼吸引力,廠商於是另外創造出「促進修復」、「延緩老化」等功能,邱品齊昨在再生醫學學會學術研討會上公開解析這類保養品的成分玄機,並質疑胜肽、生長因子、修復酵素等保養成分的實際效果。

邱品齊指出,胜肽、生長因子、修復酵素的相關研究,都只是體外層級的細胞測試,就是將高純度成分直接加到細胞培養皿。研究結果無法類推到這些成分的保養品應用在人體的效果,長期使用的副作用也還待確認。

例如,胜肽結構非常不穩,很容易被水解,但製成保養品勢必添加水、油、防腐劑,此時胜肽究竟變化成什麼樣子都是個問號,更難推論作用。

 

胜肽、生長因子效果待確認

至於「生長因子」,分子大、吸收不易,而且作用需謹慎調控,長期刺激細胞會有什麼結果也是未知。

萃取自藻類、細菌的各種「修復酵素」,多數宣稱可進入「基因」促進修復,事實上,保養品能滲進皮膚的比率不到1%,就算厚厚地敷上一層,頂多也只到5%,想要讓有效成分穿越人體組織直入基因,難度實在太高了。

邱品齊表示,臨床上實證較清楚的保護成分包括凡士林、乳油木果油、氧化鋅和高分子聚合膜較有效的修復成分則有玻尿酸鹽、蘆薈萃取等。他建議消費者購買前認真查看成分。

 

品名作文章醫師:誤導消費者

邱品齊批評說,現行法規不允許保養品宣稱有抗老、凍齡、再生、修補、重建等「功能」,結果不少廠商故意改在「品名」上作文章,刻意誤導消費者。他建議政府對「保養品」、「功能性保養品」、「藥妝品」、「藥品」的歸類、效用證實,都要在法規上有更明確的定義,消費者才不會看得一頭霧水。

食藥署副署長姜郁美表示,目前正在研擬修正化妝品衛生管理條例草案,歡迎各界提出建議,讓管理更臻周延。

Charlie 發表在 痞客邦 留言(0) 人氣()

140212130134-margarita-louis-dreyfus-olympique-football-entertain-feature  

The orphan who became a billionaire Russian Tsarina

Margarita Louis-Dreyfus with Vincent Labrune, president of French football club Olympique de Marseille, of which she is the owner.

Charlie 發表在 痞客邦 留言(0) 人氣()

49e22edb40668e2934d2d85ba1a2f071  

成功公司的創辦人有哪些特質?

作者:Sam Altman

圖片來源:SalFalko

 

本文作者

Sam Altman曾被《美國商業週刊》評為最優秀的年輕企業家,他是地理定位服務Loopt的共同創辦人和CEO,該公司於20123月被預付現金卡業務公司Green Dot收購。

 

我最近一直在思考這樣一個問題:那些非常成功的公司在剛起步的時候都是怎麼做的?從我的個人經驗出發,我列出了成功公司的創辦人所具備的十多個特徵。當然現實生活肯定有很多例外,然而很多最終沒有成功的創業公司一開始也做了這些事情,我想可能是價值與公司模式適配度上的問題。

他們在產品品質或者使用者經驗上幾近痴迷──他們在細節上花費大量時間,而且有些細節乍看似乎不是很重要。如果產品的某個部分有一點小問題,或者使用者有一個不好的經驗,這些公司的創辦人就感到渾身不舒服雖然他們堅信應該要儘快推出產品和疊代,但他們一般不會推出一款陽春的東西。(當然,這不是拖延產品發佈的藉口,不過,會不會有可能是,你為了發佈產品,花了太長了的時間?)

此外,他們不會在創辦人和使用者之間安插任何人做中介。這些公司的創辦人通常親自擔任銷售和客戶支援的工作人員

他們對人才非常痴迷成功創辦人對自己團隊的素質引以為傲,並不惜一切代價吸引人才加盟。就像人們說的一樣,他們只希望雇用最好的人,而最出色的創業家在這一點上絲毫不能妥協。如果他們在聘用人才上犯了錯誤,他們會馬上更正。

他們招募人才的過程非常緩慢。為招人而招人的話,他們壓根提不起勁去做

當然,這也說明了他們對公司文化的重視程度。

他們可以用幾個清晰的字眼來說明公司的願景。這與有些公司需要數個複雜句子來說明願景,形成鮮明的對比,而複雜的說明似乎從來沒有發揮很好的效果

同時,他們可以有條不紊的解釋為什麼他們將會成功,即使其他人在遇到某些問題後慘遭失敗他們對市場有著敏鋭的洞察力,並相信他們會掌握一個相當不錯的市場

更概括性地說,他們的溝通能力也相當不錯

他們往往很早就有賺錢的能力通常只要他們獲得了第一個使用者,就能產生收益

他們性格堅韌、冷靜。那些成功公司的創辦人都有種堅韌不拔,處事冷靜的品格。每一次運作遭遇困難瀕臨倒閉,有時甚至一天之內發生數次這種情形,他們應對這種情況就像是掏出一桿槍射擊壞人一樣,從來不會失去他們的理智。

堅韌的品格是可以挖掘和養成的,我見過一些一開始並不那麼堅強的創始人,後來很快就練就了這種品質。

他們會控制開支。除了在招募人才上很緩慢,他們在起步的時候都非常節儉。有趣的是,那些不這麽做的公司(通常都失敗了)總是理直氣壯地說:「我們在構想一個偉大的藍圖」。在一切都運行妥當之後,他們有時也會增加資金投入,但是只花在真正重要的事情上面。

他們會做一些核心使用者真正喜愛的東西Paul Buchheit是我所知道的第一個提出這個觀點的人,確實有道理。成功的新創公司幾乎總是先從使用者喜愛的產品作為最初的創業核心,然後使他們對產品產生依賴,再逐漸發展壯大。以我的經驗來看,以大多數人的喜好為出發點制定產品戰略通常都不會發揮很好的作用

他們有組織地成長。他們對於大的合作夥伴關係一般抱持懷疑態度,並盡可能在較小程度上依靠公關。他們當然不會利用大型新聞會來宣傳其創業項目。平庸的創業者才會依賴於大型公關來幫他們解答自己的成長歷程

他們專注於成長。成功的創辦人們對使用者和收益的數字了然於胸,當你問他們的時候,他們可以脫口而出。同時,他們會設定下一星期,下一個月和下一年的目標。

他們會在企業發展及未來的戰略之間做好平衡。他們有明確的計劃,並且很清楚自己要做的是什麽。但在那一刻,他們更注重的是計劃的執行,而不是制定好幾年的戰略計劃。

這一特點還可以從第一個創業項目的規模來體現。你不可能從零基礎就直接到一個大規模項目,你必須先從一個不會太大,但也不會太小的項目開始。他們似乎就有一種與生俱來的天賦,做出合適規模的項目

他們做的事情,最初都不起眼Paul Graham 已經討論過這一點。最出色的創業者通常能把小事情做得很大

他們有一種什麽事都願意做的態度。在創業過程中會有一些枯燥乏味的事情,平庸的創辦人聘僱他人來做自己不想做的事情,而出色的創辦人則會做任何他們認為是對公司有利的事情,即使他們不喜歡做

他們善於針對事情的輕重緩急安排優先次序。隨便哪一天可能有100件事情要處理。那些真正成功的創業家在這方面稱得上果決,他們要確保每天優先完成兩到三個重點工作(優先級的判斷也當然也是必須的),並忽略其他項目。

創辦人都很友好。當然並非個個如此,但我認識的最成功的創辦人都比普通人還更友好些。作為創辦人的他們,堅韌、有競爭力,有時也稱的上殘忍,但總的來說,他們都很友好

他們對假裝創建一家公司沒有興趣。他們關心的是成功,而不是「看起來成功」。他們不會因為有一個公司的實體而感到興奮,他們不會花很多時間與律師或會計師會面,或進行社交活動或其他類似的事情。他們想贏,但不在意他們看起來怎麽樣

一個極為重要的原因是,他們願意上手去做那些看似微不足道的事情,比如創辦一個網站,讓你能在別人家的氣墊床上過夜——Airbnb 就是這麽來的。最好的點子在剛起步的時候,大都備受質疑。而如果你注重表象甚於本質,你就會在意別人是否會嘲笑你。兩個選擇:一是做一家會遭人嘲笑,但迅速茁壯的公司;二是待有像有樣的漂亮辦公室裡,但公司的增長曲線是那麽的平緩。你要怎麽選?

他們對細小的事情也一絲不茍在平庸的創辦人在誇誇其談其宏偉計劃的時候,優秀的創辦人可能正在做一些看起來很小的事情,而且他們可以快速的完成這些事情。你每次跟他們談話的時候,他們可能已經完成了一些新的東西即使他們正在做一個大項目,他們也是一個一個小事件逐次攻破,並最後取得大的突破。他們永遠不會消失了一年,什麽也沒幹,然後突然間就完成了一個大項目。所以他們是值得信賴的,如果他們告訴你,他們想做點什麽,就一定會去做。

他們會迅速採取行動。對於一切事情,他們會迅速做出決定,對電子郵件也都迅速回覆。這是優秀的創辦人與平庸的創辦人之間最顯著的區別之一。優秀的創辦人都是行動機器

Charlie 發表在 痞客邦 留言(0) 人氣()

17649  

馬雲:「員工離職原因,兩點最真實:錢沒給到位、心委屈了!這些歸根到底就一條:幹得不爽!」

 

馬雲說:員工的離職原因林林總總,只有兩點最真實:

錢,沒給到位;

心,委屈了

這些歸根到底就一條:幹得不爽

 

員工臨走還費盡心思找靠譜的理由,就是為給你留面子,不想說穿你的管理有多爛、他對你已失望透頂。仔細想想,真是人性本善。作為管理者,定要樂於反省

 

帶團隊,你得問自己,人為什麼要跟著你混?

帶團隊做好這8

授人以魚:給員工養家糊口的錢。

授人以漁:教會員工做事情的方法和思路;

授人以欲:激發員工上進的欲望,讓員工樹立自己的目標;

授人以娛:把快樂帶到工作中,讓員工獲得幸福

授人以愚:告訴團隊做事情扎實、穩重,大智若愚,不可走捷徑和投機取巧。

授人以遇:給予創造團隊成長,學習,發展的機遇,成就人生。

授人以譽:幫助團隊成員獲得精神層面的讚譽,為成為更有價值的人而戰,光宗耀祖

授人以宇:上升到靈魂層次,頓悟宇宙運行智慧,樂享不惑人生

 

一流管理者:自己不幹,下屬快樂的幹;

二流管理者:自己不幹,下屬拼命的幹;

三流管理者:自己不幹,下屬主動的幹;

四流管理者:自己幹,下屬跟著幹;

五流管理者:自己幹,下屬沒事幹;

末流管理者:自己幹,下屬對著幹。

 

九段管理者修煉

一段:以身作則,堪為榜樣。

二段:幫助下屬,無私奉獻。

三段:教化下屬,為人師表。

四段:建立規則,打造團隊。

五段:高效激勵,領導思維。

六段:全面統籌,科學管理。

七段:運籌帷幄,決勝千里。

八段:機制勵人,文化凝人。

九段:組織制勝,天長地久。

 

怎麼樣留住人才?

必須給員工4個機會

做事的機會

賺錢的機會

成長的機會

發展的機會

 

必須經營員工4個感覺:

目標感,

安全感,

歸屬感,

成就感!

 

什麼是培養人才?

敢於給員工做事的機會,也能給員工犯錯的機會,這才是培養人才

 

什麼是人才?做得了事,吃得了虧,負得了責就是人才!

什麼是領導?指引得了方向,給得了方法,凝聚得了人心就是領導!

什麼是使命?活下來是為了事業,能把命都使上去就是使命!

Charlie 發表在 痞客邦 留言(0) 人氣()