Ponad 7000 publikacji medycznych!
Statystyki za 2021 rok:
odsłony: 8 805 378
Artykuły w Czytelni Medycznej o SARS-CoV-2/Covid-19

Poniżej zamieściliśmy fragment artykułu. Informacja nt. dostępu do pełnej treści artykułu
© Borgis - Postępy Nauk Medycznych 5/2014, s. 352-356
*Tomasz Skirecki1, 2, Grażyna Hoser2, Urszula Zielińska-Borkowska1
Czy mezenchymalne komórki macierzyste są szansą na przełom w leczeniu ARDS?
Are mesenchymal stem cells a chance for the breakthrough in the treatment of acute respiratory distress syndrome?
1Department of Anesthesiology and Intensive Care, Medical Center of Postgraduate Education, Warszawa
Head of Departament, a.i.: Małgorzata Malec-Milewska, MD, PhD
2Laboratory of Flow Cytometry, Medical Centrer of Postgraduate Education, Warszawa
Head of Laboratory: Grażyna Hoser, PhD
Streszczenie
Zespół ostrej niewydolności oddechowej (ang. acute respiratory distress syndrome – ARDS) jest stanem klinicznym obejmującym ciężkie i ostre uszkodzenie płuc o różnej etiologii. Zespół ten charakteryzuje się nadmierną reakcją zapalną w płucach prowadzącą do niszczenia bariery śródbłonkowo-nabłonkowej, powstania nacieków neutrofilowych, błon hialinowych, obrzęku i zmniejszenia podatności płuc i hipoksji. Wysoka śmiertelność i niepowodzenia dotychczasowych prób klinicznych z wykorzystaniem potencjalnych leków skłaniają do poszukiwania nowych opcji terapeutycznych działających wielokierunkowo. Spore nadzieje w tej mierze wiąże się z mezenchymalnymi komórkami macierzystymi (ang. mesenchymal stem cells – MSCs). Komórki te, występujące w dorosłym organizmie, mają zdolność do różnicowania się w dojrzałe komórki tkanki łącznej, a także do modulowania wrodzonej i nabytej odpowiedzi immunologicznej. Ze względu na te właściwości mogą się okazać przydatne w leczeniu ARDS. W ostatnich latach wykazano na podstawie badań różnych modeli ARDS, że podanie macierzystych komórek może wyciszać reakcję zapalną, redukować uszkodzenie płuc, a jednocześnie poprawiać zdolność do zwalczania infekcji, co razem powoduje zmniejszenie śmiertelności zwierząt z ARDS. W poniższym artykule przedstawiamy właściwości MSCs mogące mieć znaczenie terapeutyczne w odniesieniu do ARDS, a także dokonujemy przeglądu badań przedklinicznych wykorzystujących MSCs w leczeniu tego zespołu. Ostatecznie rozważamy również potencjalne trudności, jakie może napotkać wprowadzenie MSCs do praktyki klinicznej.
Summary
Acute respiratory distress syndrome (ARDS) is a clinical condition relating to a group of severe and acute lung injuries of different etiology. The syndrome is characterized by an extensive local lung inflammation leading to the destruction of the endothelial-epithelial barrier, neutrophils infiltration, formation of hyaline membranes, edema, reduction of compliance and hypoxia. High mortality rates and the failures of the clinical trials up to date with potential drugs encourages an intensive search for a new innovative pleiotropic therapeutic regimen. Mesenchymal stem cells (MSCs) constitute a population of cells with the ability to differentiate into lineages of the connective tissue and also possess the capacity to modulate both innate and adaptive immune response. These features of MSCs have justified their experimental use in the models of ARDS. In the last years it has been shown in different models that the administration of MSCs can reduce the inflammatory response, decrease lung injury but enhance the antibacterial regimens, altogether leading to the reduction of mortality. In this article we present the features of MSCs that may have an impact on the course of ARDS and we review the preclinical trials of MSCs in this syndrome. We conclude by discussing potential difficulties and challenges that can arise during the introduction of MSCs into the clinical treatment of ARDS.



ARDS – constant challange
Acute respiratory distress syndrome (ARDS) is the most severe form of lung injury with mortality reaching up to 40% (1). It is also one of the most common clinical syndromes treated in the intensive care units (ICU). The etiology of ARDS is very broad and associated with multiple other conditions. Clasically, two sources of ARDS are distinguished: pulmonary (i.e. pneumonia, choking, inhalatory burn) and non-pulmonary sources (sepsis, acute pancreatitis, trauma, massive blood transfusions). The most frequent cause of ARDS is sepsis (2). The diagnostic proces is based on clinical signs like: acute beginning, hypoxia (PaO2/FiO2 < 300), bilateral opacities on chest imaging not explained by other pulmonary pathology (e.g. pleural effusion, pneumothorax, or nodules) and respiratory failure not explained by heart failure or volume overload (3). Heterogenous clinical picture and very wide range of patients together with lack of specific symptoms constitute the first therapeutical problem. Although the pathomorphological changes in ARDS are relatively well defined (presence of hyalin membranes, neutrophil infiltrates, significant alveolar damage followed by a pattern of fibrosis and regeneration), the pathophysiological processes are complicated and remain not well understood. The first, acute phase of ARDS is characterized by exaggerated inflammatory response reflected by e.g. increased levels of pro-inflammatory cytokines: TNF, IL-6, IL-8 in serum and bronchoalveolar lavage fluid, activation of pulmonary macrophages and infiltration of activated neutrophils. These processes along with factors inducing ARDS (e.g. bacterial toxins, hot gases) evoke apoptosis of pneumocytes and injury of the microcirculatory endothelium of lungs. The injury of endothelial-epithelial barrier enhances further influx of neutrophils and is a cause of pulmonary edema (4, 5). Even in the early phase of the disease, simultaneously with the inflammatory process, activation of fibroblasts and regenerative mechanisms begins (6). Nowadays, patients rarely die because of the early respiratory failure. Common cause of deaths include nosocomial infections and development of Multiorgan failure. In 50% of patients who died with ARDS, lung fibrosis is observed, what suggests impaired activation of regenerative mechanisms what causes deterioration of clinical state (7).
In spite of many clinical trials and many years of intensive research, there is no effective treatment of ARDS so far. Conducted trials with glucocorticoids, exogenous surfactant, inhaled nitric oxide, prostaglandins, anticoagulants did not improve the outcome (8). Reduction of mortality was only achieved by introducing protective mechanical ventilation (airway pressure not exceeding 30 cm H2O), restrictive fluid therapy and ventilation in prone position (9-11). These procedures rather limit the iatrogenic injury than treat the disease. Due to the complex pathophysiology and harmful influence of injured lungs on other organs, effective treatment of ARDS should be multidirectional.
Mesenchymal stem cells – unique features pave the way
Originally, mesenchymal stromal cells (called also mesenchymal stem cells) have been isolated and characterized from the bone marrow by Friedenstein et al. (12). These multipotent stem cells have the capacity to multilineage differentiation into the cells of the connective tissue and therefore play an important role in the regenerative mechanisms of the adult organism. Other research groups have identified these cells in virtually every organ of the human body (lungs, heart, liver, adipose tissue, placenta, cord blood) (13). Localization among the pericytes surrounding vessels present in all tissues explain nicely the widespread presence of these cells in the body (14). Primary and still the most important method of isolation of MSCs is a culture of adherent cell colonies from a given tissue. Although, a consensus was made about the methods that confirm presence of MSCs, there is a lack of efficient method of their isolation based on one antigen. Such method is needed to obtain a homogenous population of cells. Criteria of MSCs are fulfilled by cells that are able to form adherent colonies in the in vitro culture, under special medium can differentiate into osteoblasts, adipocytes and chondrocytes and finally they express surface markers as: CD90, CD105, CD73, CD44 but not: CD45, CD34, CD31 (15).
MSCs are in the focus of interest of investigators working on the new therapies of multiple diseases (e.g. graft versus host disease, inflammatory bowel disease, infarction, multiple sclerosis) (16). MSCs seem so attractive because of their capacities to modulate the immune response and injured tissues, although their potential to differentiate into harmed tissues is also important. First reports on the utility of MSCs in the regeneration of injured lungs, suggesting high level of engraftment by transplanted cells (17), were not confirmed by other groups (18-21). In these reports, the level of engraftment in the injured lungs was below 1% of lung cells what suggests other mechanisms of action of injected MSCs. Currently, many research are aimed at investigating the immunomodulatory properties of MSCs. Numerous papers reported immunosuppressive effects of these cell on both innate and adaptive immunity (22-25). This effect is achieved by direct contact between cells and also by paracrine mediators. MSCs produce factors like: PGE2, indoleamine 2,3-dioxygenase, cytokines: TGFβ, IL-1RA, IL-10. The role of PGE2 was widely investigated and described: produced by MSCs – “re-programs” pulmonary macrophages during sepsis to produce IL-10 (26). IL-10 is a key mediator in the protective pathways utilized by transplanted MSCs in the organ injury models in sepsis (26). In the animal model of direct lung injury by endotoxin with subsequent intratracheal application of MSCs, the reduction of mortality and improvement of pathomorphological picture of lungs was correlated with reduced concentration of TNF and increased level of IL-10 in the BALF and serum (27). However, the interaction between MSCs and immunity is more complexed. These cells can also improve the anti-microbial defense what is extremely important in the sepsis induced ARDS and in the prevention of nosocomial infections in sterile ARDS. After stimulation with TNF, MSCs secrete IL-6 (28), which was shown in in vitro models to induce production of IgG by B cells (29). Also, an effect of MSCs on granulocytes is very interesting. MSCs inhibit apoptosis and degranulation of granulocytes, improving their phagocytic capacity (30). MSCs also secrete anti-bacterial peptide called LL-37 (31), what makes themselves a part of the immune system. Abovementioned interactions seem however, quite selective knowing the results of studies examining effect of MSCs on the pattern of genes expression in a murine model of sepsis. The study has revealed that systemic application of MSCs re-programs expression of hundreds of immune-related genes (32).

Powyżej zamieściliśmy fragment artykułu, do którego możesz uzyskać pełny dostęp.
Mam kod dostępu
  • Aby uzyskać płatny dostęp do pełnej treści powyższego artykułu albo wszystkich artykułów (w zależności od wybranej opcji), należy wprowadzić kod.
  • Wprowadzając kod, akceptują Państwo treść Regulaminu oraz potwierdzają zapoznanie się z nim.
  • Aby kupić kod proszę skorzystać z jednej z poniższych opcji.

Opcja #1

24

Wybieram
  • dostęp do tego artykułu
  • dostęp na 7 dni

uzyskany kod musi być wprowadzony na stronie artykułu, do którego został wykupiony

Opcja #2

59

Wybieram
  • dostęp do tego i pozostałych ponad 7000 artykułów
  • dostęp na 30 dni
  • najpopularniejsza opcja

Opcja #3

119

Wybieram
  • dostęp do tego i pozostałych ponad 7000 artykułów
  • dostęp na 90 dni
  • oszczędzasz 28 zł
Piśmiennictwo
1. Brun-Buisson C, Minelli C, Bertolini G et al.: Epidemiology and outcome of acute lung injury in European intensive care units. Results from the ALIVE study. Intensive Care Med 2004; 30: 51-61.
2. Sheu CC, Gong MN, Zhai R et al.: Clinical characteristics and outcomes of sepsis-related vs non-sepsis-related ARDS. Chest 2010; 138: 559-567.
3. ARDS Definition Task Force, Ranieri VM, Rubenfeld GD et al.: Acute respiratory distress syndrome: the Berlin Definition. JAMA 2012; 307: 2526-2533.
4. Bhatia M, Moochhala S: Role of inflammatory mediators in the pathophysiology of acute respiratory distress syndrome. J Pathol 2004; 202: 145-156.
5. Ware LB: Pathophysiology of acute lung injury and the acute respiratory distress syndrome. Semin Respir Crit Care Med 2006; 27: 337-349.
6. Rocco PR, Dos Santos C, Pelosi P: Lung parenchyma remodeling in acute respiratory distress syndrome. Minerva Anestesiol 2009; 75: 730-734.
7. Martin C, Papazian L, Payan MJ et al.: Pulmonary fibrosis correlates with outcome in adult respiratory distress syndrome. A study in mechanically ventilated patients. Chest 1995; 107: 196-200.
8. Saguil A, Fargo M: Acute respiratory distress syndrome: diagnosis and management. Am Fam Physician 2012; 85: 352-358.
9. Roche-Campo F, Aguirre-Bermeo H, Mancebo J: Prone positioning in acute respiratory distress syndrome (ARDS): when and how? Presse Med 2011; 40: e585-594.
10. The Acute Respiratory Distress Syndrome Network. Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome. N Engl J Med 2000; 342: 1301-1308.
11. Wiedemann HP, Wheeler AP, Bernard GR et al.: Comparison of two fluid-management strategies in acute lung injury. N Engl J Med 2006; 354: 2564-2575.
12. Friedenstein AJ, Petrakova KV, Kurolesova AI, Frolova GP: Heterotopic of bone marrow. Analysis of precursor cells for osteogenic and hematopoietic tissues. Transplantation 1968; 6: 230-247.
13. Williams AR, Hare JM: Mesenchymal stem cells: biology, pathophysiology, translational findings, and therapeutic implications for cardiac disease. Circ Res 2011; 109: 923-940.
14. Corselli M, Chen CW, Crisan M et al.: Perivascular ancestors of adult multipotent stem cells. Arterioscler Thromb Vasc Biol 2010; 30: 1104-1109.
15. Dominici M, Le Blanc K, Mueller I et al.: Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006; 8: 315-317.
16. Trounson A, Thakar RG, Lomax G et al.: Clinical trials for stem cell therapies. BMC Med 2011; 9: 52.
17. Krause DS, Theise ND, Collector MI et al.: Multi-organ, multi-lineage engraftment by a single bone marrow-derived stem cell. Cell 2001; 105: 369-377.
18. Kotton DN, Ma BY, Cardoso WV et al.: Bone marrow-derived cells as progenitors of lung alveolar epithelium. Development 2001; 128: 5181-5188.
19. Ortiz LA, Gambelli F, McBride C et al.: Mesenchymal stem cell engraftment in lung is enhanced in response to bleomycin exposure and ameliorates its fibrotic effects. Proc Natl Acad Sci U S A 2003; 100: 8407-8411.
20. Kotton DN, Fabian AJ, Mulligan RC: Failure of bone marrow to reconstitute lung epithelium. Am J Respir Cell Mol Biol 2005; 33: 328-334.
21. Loi R, Beckett T, Goncz KK et al.: Limited restoration of cystic fibrosis lung epithelium in vivo with adult bone marrow-derived cells. Am J Respir Crit Care Med 2006; 173: 171-179.
22. Gebler A, Zabel O, Seliger B: The immunomodulatory capacity of mesenchymal stem cells. Trends Mol Med 2012; 18: 128-134.
23. Zhang W, Ge W, Li C et al.: Effects of mesenchymal stem cells on differentiation, maturation and function of human monocyte-derived dendritic cells. Stem Cells Dev 2004; 13: 263-271.
24. Di Nicola M, Carlo-Stella C, Magni M et al.: Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood 2002; 99: 3838-3843.
25. Spaggiari GM, Capobianco A, Abdelrazik H et al.: Mesenchymal stem cells inhibit natural killer-cell proliferation, cytotoxicity, and cytokine production: role of indoleamine 2,3-dioxygenase and prostaglandin E2. Blood 2008; 111: 1327-1333.
26. Nèmeth K, Leelahavanichkul A, Yuen PS et al.: Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat Med 2009; 15: 42-49.
27. Gupta N, Su X, Popov B et al.: Intrapulmonary delivery of bone marrow-derived mesenchymal stem cells improves survival and attenuates endotoxin-induced acute lung injury in mice. J Immunol 2007; 179: 1855-1863.
28. van den Berk LC, Jansen BJ, Siebers-Vermeulen KG et al.: Mesenchymal stem cells respond to TNF but do not produce TNF. J Leukoc Biol 2010; 87: 283-289.
29. Rasmusson I, Le Blanc K, Sundberg B et al.: Mesenchymal stem cells stimulate antibody secretion in human B cells. Scand J Immunol 2007; 65: 336-343.
30. Raffaghello L, Bianchi G, Bertolotto M et al.: Human mesenchymal stem cells inhibit neutrophil apoptosis: a model for neutrophil preservation in the bone marrow niche. Stem Cells 2008; 26: 151-162.
31. Krasnodembskaya A, Song Y, Fang X et al.: Antibacterial effect of human mesenchymal stem cells is mediated in part from secretion of the antimicrobial peptide LL-37. Stem Cells 2010; 28: 2229-2238.
32. Mei SH, Haitsma JJ, Dos Santos CC et al.: Mesenchymal stem cells reduce inflammation while enhancing bacterial clearance and improving survival in sepsis. Am J Respir Crit Care Med 2010; 182: 1047-1057.
33. Nemzek JA, Ebong SJ, Kim J et al.: Keratinocyte growth factor pretreatment is associated with decreased macrophage inflammatory protein-2alpha concentrations and reduced neutrophil recruitment in acid aspiration lung injury. Shock 2002; 18: 501-506.
34. Sugahara K, Iyama K, Kuroda MJ, Sano K: Double intratracheal instillation of keratinocyte growth factor prevents bleomycin-induced lung fibrosis in rats. J Pathol 1998; 186: 90-98.
35. Guery BP, Mason CM, Dobard EP et al.: Keratinocyte growth factor increases transalveolar sodium reabsorption in normal and injured rat lungs. Am J Respir Crit Care Med 1997; 155: 1777-1784.
36. Birukova AA, Alekseeva E, Mikaelyan A, Birukov KG: HGF attenuates thrombin-induced endothelial permeability by Tiam1-mediated activation of the Rac pathway and by Tiam1/Rac-dependent inhibition of the Rho pathway. FASEB J 2007; 21: 2776-2786.
37. Patel SA, Sherman L, Munoz J, Rameshwar P: Immunological properties of mesenchymal stem cells and clinical implications. Arch Immunol Ther Exp (Warsz) 2008; 56: 1-8.
38. Matute-Bello G, Frevert CW, Martin TR: Animal models of acute lung injury. Am J Physiol Lung Cell Mol Physiol 2008; 295: L379-399.
39. Lee JW, Fang X, Gupta N et al.: Allogeneic human mesenchymal stem cells for treatment of E. coli endotoxin-induced acute lung injury in the ex vivo perfused human lung. Proc Natl Acad Sci U S A 2009; 106: 16357-16362.
40. Rojas M, Xu J, Woods CR et al.: Bone marrow-derived mesenchymal stem cells in repair of the injured lung. Am J Respir Cell Mol Biol 2005; 33: 145-152.
41. Xu J, Qu J, Cao L et al.: Mesenchymal stem cell-based angiopoietin-1 gene therapy for acute lung injury induced by lipopolysaccharide in mice. J Pathol 2008; 214: 472-481.
42. Moodley Y, Atienza D, Manuelpillai U et al.: Human umbilical cord mesenchymal stem cells reduce fibrosis of bleomycin-induced lung injury. Am J Pathol 2009; 175: 303-313.
43. Danchuk S, Ylostalo JH, Hossain F et al.: Human multipotent stromal cells attenuate lipopolysaccharide-induced acute lung injury in mice via secretion of tumor necrosis factor-α-induced protein 6. Stem Cell Res Ther 2011; 2: 27.
44. Chien MH, Bien MY, Ku CC et al.: Systemic human orbital fat-derived stem/stromal cell transplantation ameliorates acute inflammation in lipopolysaccharide-induced acute lung injury. Crit Care Med 2012; 40: 1245-1253.
45. Curley GF, Hayes M, Ansari B et al.: Mesenchymal stem cells enhance recovery and repair following ventilator-induced lung injury in the rat. Thorax 2012; 67: 496-501.
46. Li J, Li D, Liu X et al.: Human umbilical cord mesenchymal stem cells reduce systemic inflammation and attenuate LPS-induced acute lung injury in rats. J Inflamm (Lond) 2012; 9: 33.
47. Brandau S, Jakob M, Hemeda H et al.: Tissue-resident mesenchymal stem cells attract peripheral blood neutrophils and enhance their inflammatory activity in response to microbial challenge. J Leukoc Biol 2010; 88: 1005-1015.
48. Kuzmina LA, Petinati NA, Parovichnikova EN et al.: Multipotent Mesenchymal Stromal Cells for the Prophylaxis of Acute Graft-versus-Host Disease-A Phase II Study. Stem Cells Int 2012; 2012: 968213.
49. Jiang R, Han Z, Zhuo G et al.: Transplantation of placenta-derived mesenchymal stem cells in type 2 diabetes: a pilot study. Front Med 2011; 5: 94-100.
50. Perin EC, Silva GV, Henry TD et al.: A randomized study of transendocardial injection of autologous bone marrow mononuclear cells and cell function analysis in ischemic heart failure (FOCUS-HF). Am Heart J 2011; 161: 1078-1087.
51. Williams AR, Trachtenberg B, Velazquez DL et al.: Intramyocardial stem cell injection in patients with ischemic cardiomyopathy: functional recovery and reverse remodeling. Circ Res 2011; 108: 792-796.
52. Bhasin A, Srivastava MV, Kumaran SS et al.: Autologous mesenchymal stem cells in chronic stroke. Cerebrovasc Dis Extra 2011; 1: 93-104.
otrzymano: 2014-02-19
zaakceptowano do druku: 2014-03-26

Adres do korespondencji:
*Tomasz Skirecki
Department of Anesthesiology and Intensive Care Medical Center of Postgraduate Education
ul. Czerniakowska 231, 00-416 Warszawa
tel. +48 (22) 584-1-220
tskirecki@gmail.com

Postępy Nauk Medycznych 5/2014
Strona internetowa czasopisma Postępy Nauk Medycznych