In this issue of Blood, Di Buduo et al report an artificial scaffold of the bone marrow niche whereby Bombyx mori silkworm cocoons appeared to coordinate several factors required for megakaryopoiesis and platelet biogenesis.1 

Di Buduo et al demonstrated a combined model to mimic the bone marrow environment to study thrombopoiesis. They previously proposed that silk material-coated microtube structure is capable of recapitulating bone marrow environment, but it failed to obtain a higher rate of mature Mks.8  The improved system included increasing stiffness, extracellular matrix, cytokines, endothelial cells, and modulating shear stress by red blood cells. In this article,1  the authors developed the 3D bioreactor that enhances migration and maturation of Mks, proplatelet generation, and yield of functional platelets. Silk material may be suitable for integration of several factors required for thrombopoiesis ex vivo. Adapted from Figures 5, 6, and 7 in the article by Di Buduo et al beginning on page 2254.

Di Buduo et al demonstrated a combined model to mimic the bone marrow environment to study thrombopoiesis. They previously proposed that silk material-coated microtube structure is capable of recapitulating bone marrow environment, but it failed to obtain a higher rate of mature Mks.8  The improved system included increasing stiffness, extracellular matrix, cytokines, endothelial cells, and modulating shear stress by red blood cells. In this article,1  the authors developed the 3D bioreactor that enhances migration and maturation of Mks, proplatelet generation, and yield of functional platelets. Silk material may be suitable for integration of several factors required for thrombopoiesis ex vivo. Adapted from Figures 5, 6, and 7 in the article by Di Buduo et al beginning on page 2254.

Close modal

A single bone marrow megakaryocyte (Mk) in the body produces >2000 platelets.2  By recapitulating the chemical and physical signaling that promotes Mk differentiation, maturation, and the release of proplatelets and platelets into the blood stream in vivo, experimental trials ex vivo have tried but failed to achieve similar numbers.3,4  Building on these ex vivo systems have been designs that incorporate switching the culture from 2-dimensional to 3-dimensional (3D) culture environments, for example, using scaffolds made of polyester fabric, hydrogel, or polydimethylsiloxane (PDMS) along with cytokines, extracellular matrix, or endothelial cells.5-7  3D environments likely make for more surface area, which could permit more proplatelets to interact with the endothelial wall, increasing the number of platelets acquired.

A series of bioreactor systems that better mimic the bone marrow environment by including extracellular matrix components, surface topography, stiffness, cytokines, and shear stress is also being investigated. Di Buduo et al,1  to obtain higher production efficiency of functional platelets from cultured Mks, prepared a bioreactor that uses a silk sponge, whereby a silk sponge-coated tube structure made by PDMS is also covered by endothelial cells or by vascular endothelial growth factor and vascular cell adhesion molecule-1 on the inside of the tube under the presence of shear stress. The idea of using the silk sponge was previously proposed by the same authors’ group.8  Di Buduo et al added additional factors necessary to fully recapitulate obtaining “functional platelets” through the artificial bone marrow environment (see figure). They previously designed a bioreactor that included both the artificial osteoblast and perivascular niches with the appropriate extracellular matrix proteins and growth factors. However, there were very few Mks displaying proplatelets and producing platelets with impaired function.8  An advance by Di Buduo et al is the incorporation of various factors, ie, composed of stiffness for Mk adhesion, various extracellular matrixes, coated endothelial cells, and an intervention by red blood cells (RBCs) to modulate flow viscosity and shear stress. The subsequent flow by RBCs might influence the conditions of the silk sponge. The silk sponge should control the elements required for mimicking a true bone marrow environment. Thus, the authors concluded that silkworm cocoons contribute to making a comfortable and flexible environment for megakaryopoiesis and platelet biogenesis, which the authors referred to as a “programmable 3D silk bone marrow niche” (see figure).1 

Because shear stress has been found to be an important factor in platelet number, Thon et al manufactured a microfluidic bioreactor that also incorporates bone marrow stiffness, the extracellular matrix composition, and other factors modulating shear stress.7  They demonstrated that best rate of shear stress resulted in Mk maturation, as proplatelets were observed within seconds of trapping compared with the several hours seen in static conditions. The shear stress in their system was generated by parallel flows.7  Nakagawa et al found, however, that a confluent system may be more effective, as flow intersecting at 60° achieves a 3.6-fold increase compared with a single-flow system.6  Therefore, a silk-based sponge with PDMS may generate more complicated shear stress conditions by preincubating with RBCs. It has been reported that shear stress increases the expression of RUNX1 in CD41a+ cells,9  also suggesting that shear stress per se facilitates megakaryopoiesis.

Ex vivo platelet production is still far from an industrial scale in transfusion medicine or a drug delivery system, but the bioreactor does offer an excellent model to study megakaryopoiesis from hematopoietic stem cells (HSCs), as demonstrated by the findings that HSCs and Mks are very close in proximity in the hematopoiesis hierarchy in the bone marrow.10 

Conflict-of-interest disclosure: The author declares no competing financial interests.

1
Di Buduo
 
CA
Wray
 
LS
Tozzi
 
L
, et al. 
Programmable 3D silk bone marrow niche for platelet generation ex vivo and modeling of megakaryopoiesis pathologies.
Blood
2015
, vol. 
125
 
14
(pg. 
2254
-
2264
)
2
Avanzi
 
MP
Mitchell
 
WB
Ex vivo production of platelets from stem cells.
Br J Haematol
2014
, vol. 
165
 
2
(pg. 
237
-
247
)
3
Matsunaga
 
T
Tanaka
 
I
Kobune
 
M
, et al. 
Ex vivo large-scale generation of human platelets from cord blood CD34+ cells.
Stem Cells
2006
, vol. 
24
 
12
(pg. 
2877
-
2887
)
4
Takayama
 
N
Nishimura
 
S
Nakamura
 
S
, et al. 
Transient activation of c-MYC expression is critical for efficient platelet generation from human induced pluripotent stem cells.
J Exp Med
2010
, vol. 
207
 
13
(pg. 
2817
-
2830
)
5
Sullenbarger
 
B
Bahng
 
JH
Gruner
 
R
Kotov
 
N
Lasky
 
LC
Prolonged continuous in vitro human platelet production using three-dimensional scaffolds.
Exp Hematol
2009
, vol. 
37
 
1
(pg. 
101
-
110
)
6
Nakagawa
 
Y
Nakamura
 
S
Nakajima
 
M
, et al. 
Two differential flows in a bioreactor promoted platelet generation from human pluripotent stem cell-derived megakaryocytes.
Exp Hematol
2013
, vol. 
41
 
8
(pg. 
742
-
748
)
7
Thon
 
JN
Mazuitis
 
L
Wu
 
S
, et al. 
Platelet bioreactor-on-a-chip.
Blood
2014
, vol. 
124
 
12
(pg. 
1857
-
1867
)
8
Pallotta
 
I
Lovett
 
M
Kaplan
 
DL
Balduini
 
A
Three-dimensional system for the in vitro study of megakaryocytes and functional platelet production using silk-based vascular tubes.
Tissue Eng Part C Methods
2011
, vol. 
17
 
12
(pg. 
1223
-
1232
)
9
Adamo
 
L
Naveiras
 
O
Wenzel
 
PL
, et al. 
Biomechanical forces promote embryonic haematopoiesis.
Nature
2009
, vol. 
459
 
7250
(pg. 
1131
-
1135
)
10
Yamamoto
 
R
Morita
 
Y
Ooehara
 
J
, et al. 
Clonal analysis unveils self-renewing lineage-restricted progenitors generated directly from hematopoietic stem cells.
Cell
2013
, vol. 
154
 
5
(pg. 
1112
-
1126
)
Sign in via your Institution