• No results found

Modeling of Cell Proliferation with Flow Cytometry Data from CFSE-based Assays

N/A
N/A
Protected

Academic year: 2021

Share "Modeling of Cell Proliferation with Flow Cytometry Data from CFSE-based Assays"

Copied!
53
0
0

Loading.... (view fulltext now)

Full text

(1)

Modeling of Cell Proliferation with Flow

Cytometry Data from CFSE-based Assays

H.T. Banks

Center for Quantitative Sciences in Biomedicine Center for Research in Scientific Computation

Department of Mathematics North Carolina State University

(2)

Clay Thompsonand Karyn Sutton

CRSC Postdocs at NCSU

Tim Schenkel

Department of Virology, Saarland University, Homburg, Germany

Jordi Argilaguet, Sandra Giest,

Cristina Peligero, Andreas Meyerhans

ICREA Infection Biology Lab, Univ. Pompeu Fabra, Barcelona, Spain

Gennady Bocharov

(3)

1 CFSE Data Overview

2 Initial Fragmentation Model of CFSE Data

3 Model Summaries 4 Data Statistical Model 5 Results

(4)

Selected Sources and References

H.T. Banks, Frederique Charles, Marie Doumic, Karyn L. Sutton, and W. Clayton Thompson, Label structured cell proliferation models, Appl. Math. Letters 23 (2010), 1412–1415;

doi:10.1016/j.aml.2010.07.009

H.T. Banks, Karyn L. Sutton, W. Clayton Thompson, Gennady Bocharov, Dirk Roose, Tim Schenkel, and Andreas Meyerhans, Estimation of cell proliferation dynamics using CFSE data, CRSC-TR09-17, North Carolina State University, August 2009; Bull. Math. Biol., 70 (2011), 116–150.

H.T. Banks, Karyn L. Sutton, W. Clayton Thompson, G. Bocharov, Marie Doumic, Tim Schenkel, Jordi Argilaguet, Sandra Giest, Cristina Peligero, and Andreas Meyerhans, A new model for the estimation of cell proliferation dynamics using CFSE data, CRSC-TR11-05, North Carolina State University, Revised July 2011; J. Immunological Methods, 373 (2011), 143–160; DOI:10.1016/j.jim.2011.08.014.

H.T. Banks, W. Clayton Thompson, Jordi Argilaguet, Cristina Peligero, and Andreas Meyerhans, A division-dependent compartmental model for computing cell numbers in CFSE-based lymphocyte proliferation assays, CRSC-TR12-03, N. C. State University, Raleigh, NC, January, 2012; Math. Biosci. Engr., to appear.

H.T. Banks and W. Clayton Thompson, Mathematical models of dividing cell populations: Application to CFSE data, CRSC-TR12-10, N. C. State University, Raleigh, NC, April, 2012; Journal on Mathematical Modelling of Natural Phenomena, to appear.

H.T. Banks and W. Clayton Thompson, A division-dependent compartmental model with cyton and intracellular label dynamics, CRSC-TR12-12, N. C. State University, Raleigh, NC, May, 2012; Intl. J. of Pure and Applied Math., 77 (2012), 119–147.

(5)

Selected Sources and References

H.T. Banks, Amanda Choi, Tori Huffman, John Nardini, Laura Poag and W. Clayton Thompson, Modeling CFSE label decay in flow cytometry data, CRSC-TR12-20, N. C. State University, Raleigh, NC, November, 2012; Applied Mathematical Letters, (2103), doi:10.1016/j.aml.2012.12.010.

H.T. Banks, D. F. Kapraun, W. Clayton Thompson, Cristina Peligero, Jordi Argilaguet and Andreas Meyerhans, A novel statistical analysis and interpretation of flow cytometry data, CRSC-TR12-23, N. C. State University, Raleigh, NC, December, 2012; J. Biological Dynamics, submitted.

A.V. Gett and P.D. Hodgkin, A cellular calculus for signal integration by T cells, Nature Immunology 1 (2000), 239–244.

E.D. Hawkins, Mirja Hommel, M.L Turner, Francis Battye, J Markham and P.D Hodgkin, Measuring lymphocyte proliferation, survival and differentiation using CFSE time-series data, Nature Protocols, 2 (2007), 2057–2067.

E.D. Hawkins, M.L. Turner, M.R. Dowling, C. van Gend, and P.D. Hodgkin, A model of immune regulation as a consequence of randomized lymphocyte division and death times, Proc. Natl. Acad. Sci., 104 (2007), 5032–5037.

T. Luzyanina, D. Roose, T. Schenkel, M. Sester, S. Ehl, A. Meyerhans, and G. Bocharov, Numerical modelling of label-structured cell population growth using CFSE distribution data, Theoretical Biology and Medical Modelling 4 (2007), Published Online.

C. Parish, Fluorescent dyes for lymphocyte migration and proliferation studies, Immunology and Cell Biol.

(6)

Selected Sources and References

B. Quah, H. Warren and C. Parish, Monitoring lymphocyte proliferation in vitro and in vivo with the intracellular fluorescent dye carboxyfluorescein diacetate succinimidyl ester, Nature Protocols 2:9 (2007), 2049–2056.

D. Schittler, J. Hasenauer, and F. Allg¨ower, A generalized model for cell proliferation: Integrating division numbers and label dynamics, Proc. Eighth International Workshop on Computational Systems Biology (WCSB 2011), June 2001, Zurich, Switzerland, p. 165–168.

J. Hasenauer, D. Schittler and F. Allg¨ower, Analysis and simulation of division- and label-structured population models: A new tool to analyze proliferation assays, Bull. Math. Biol., 74 (2012), 2692–2732.

(7)

Scientific Goals

Long-term collaboration between NCSU and UPF Monitor cell division and differentiation

Assess polyfunctionality

Investigate immunospecific extracellular signaling pathways Identify correlated and (ideally) causal relationships

between immune mechanisms

Quantitative measure of ‘dynamic responsiveness’ Link observed (cellular) behaviors to clinical outcomes; improve clinical outcomes

The ‘central problem in immunology’ (according to G. Bocharov): to understand the ‘cellular and molecular mechanisms that control the ability of the immune system to mount a protective response against pathogen-derived foreign antigens, but avoid a pathological response to self-antigens’

(8)

Persistent Infections

Trade-off between immune protection and immunopathy can lead to persistent infection

(9)

Persistent Infections (cont’d)

(A. Meyerhans)

‘Dynamic equilibrium’ between host immune response and microbe expansion

Applications in HIV, HCV, TB Open questions:

Characterization of set point

(10)

Goals

Observations:

Clinical outcome believed to be strongly influenced by timing and magnitude of ‘clonal expansion’

CFSE (intracellular dye) + flow cytometry = powerful new tool for tracking cell division

Analysis of data:

Develop a mathematical model for CFSE data

Link cell counts to measures of proliferation/death rates Population doubling time; cell cycle time

Cell viability Applications:

Analyze experimental and biological variability Optimal experimental design

(11)

CFSE Data Set

0 0.5 1 1.5 2 2.5 3 3.5 0 0.5 1 1.5 2 2.5 3 3.5x 10

5 CFSE Time Series Histogram Data

Label Intensity z [Log UI]

Structured Population Density [numbers/UI]

t = 0 hrs t = 24 hrs t = 48 hrs t = 72 hrs t = 96 hrs t = 120 hrs

(12)

Data Overview

(13)

CFSE Labeling (Lyons and Parish, 1994)

Cells cultured with CFDA-SE (carboxyfluorescein diacetate succinimidyl ester) then washed

CFDA-SE becomes protein-bound and fluorescent CFSE (the fluorescent dye carboxyfluorescein succinimidyl ester) Dye split between daughter cells at division

Dye naturally turns over/degrades (very slowly)

Fluorescence Intensity (FI) of CFSE measured via flow cytometry

FI linear with dye concentrationFImass

(14)

CFSE Labeling (Lyons and Parish, 1994)

(C. Parish, Fluorescent dyes for lymphocyte migration and proliferation studies, Immunology and Cell Biol. 77 (1999), 499–508.)

(15)

CFSE Data Set

0 0.5 1 1.5 2 2.5 3 3.5 0 0.5 1 1.5 2 2.5 3 3.5x 10

5 CFSE Time Series Histogram Data

Label Intensity z [Log UI]

Structured Population Density [numbers/UI]

t = 0 hrs t = 24 hrs t = 48 hrs t = 72 hrs t = 96 hrs t = 120 hrs

(16)

Goals of Modeling

Cellular ‘Dynamic Responsiveness’

Link cell counts with proliferation/death rates

Population doubling time Cell viability

Biological descriptors (cell cycle time, etc.)

Uncertainty Quantification...

... in the experimental procedure ... for estimated rates/etc

Analyze cell differentiation and division-linked changes Investigate immunospecific extracellular signaling pathways

(17)

Traditional Approach (Gett and Hodgkin)

Traditional ‘semi-quantitative analysis’ pioneered by Gett and Hodgkin et al. (2000)

Fit data with gaussian curves to determine approximate cells per generation

(A.V. Gett and P.D. Hodgkin, A cellular calculus for signal integration by T cells, Nature Immunology 1 (2000), 239–244.)

(18)

Traditional Approach (cont’d)

Gett-Hodgkin method quick, easy to implement, useful comparisons between data sets (e.g. stimulation conditions)

Compatible with ODE, DDE models; ‘indirect fitting’ for parameter estimation

Generalizations, extensions, and various other modeling efforts

Smith-Martin model (with generalizations) Cyton model

(19)

Label-Structured Model

All previous work with cell numbers determined by deconvolution

Alternatively, we propose to fit the CFSE histogram data directly

Capture full behavior of the population density

No assumption on the shape of CFSE uptake/distribution

Histogram presentation of cytometry data makes

structured population modelsa natural choice (as in age,

size, etc) except here structure label is“CFSE content or

intensity”

Key ideas first formulated by Luzyanina, Bocharov, et al., 2007

(20)

Label-Structured Model (cont’d)

This model must account for (Luzyanina et al., 2007): Slow decay of CFSE FI over time

Dilution of CFSE as cells divide Asynchronous division times

0 0.5 1 1.5 2 2.5 3 3.5 0 0.5 1 1.5 2 2.5 3 3.5x 10

5 Data in Logarithmic Coordinate (z)

z [Log UI] Cell Counts t = 24 hrs t = 48 hrs t = 96 hrs t = 120 hrs

(21)

Rate of Label Decay

(C. Parish, Fluorescent dyes for lymphocyte migration and proliferation studies, Immunology and Cell Biol. 77 (1999), 499–508.)

(22)

‘Biphasic Decay’

0 20 40 60 80 100 120 140 160 1.5 2 2.5 3 3.5 4 4.5 5x 10

4 Donor 1 Exponential Fit,

Time (hrs) CFSE Intensity dx dt =v(x) =c(xxa) Exponential 0 20 40 60 80 100 120 140 160 1.5 2 2.5 3 3.5 4 4.5 5x 10

4 Donor 1 Gompertz Fit,

Time (hrs) CFSE Intensity dx dt =v(t,x) =c(xxa)e −kt Gompertz

(23)

‘Translated Coordinate’ (Banks et al, 2010)

While the structure variable z does correlate with division number, the ‘translated variable’

s(t,z) =z c

k log 10

ekt

−1

has an even stronger correlation.

Follows from the Method of Characteristics

Intuitively, s represents the FI of a cell with a hypothetical label which does not decay

(24)

‘Translated Coordinate’ (cont’d)

0 0.5 1 1.5 2 2.5 3 3.5 0 0.5 1 1.5 2 2.5 3 3.5x 10

5 Data in Logarithmic Coordinate (z)

z [Log UI] Cell Counts t = 24 hrs t = 48 hrs t = 96 hrs t = 120 hrs −10 −0.5 0 0.5 1 1.5 2 2.5 3 3.5 4 0.5 1 1.5 2 2.5 3 3.5x 10 5 X: 1.592 Y: 3.109e+05 Data in Translated Coordinate s

s [Log UI] Cell Counts t = 24 hrs t = 48 hrs t = 96 hrs t = 120 hrs

(25)

Fragmentation Model Summary

Model is capable of precisely fitting the observed data

c, k , xaestimated consistently (asαandβ nodes change),

though subject to high experimental variability ‘Translated coordinate’ very strongly correlated with division number

Time-dependence of the proliferation rate is an essential feature of the model

Biologically relevant average values of proliferation and death (in terms of number of divisions undergone) are easily computable.

But...(Aggregate Data/Aggregate Dynamics)

Still cannot compute cell numbers and cohort rate parameters

Data overlap affecting estimated rates (?) Large number of parameters necessary

(26)

Best-fit, AIC-selected results: Model A5B5Dist

0 0.5 1 1.5 2 2.5 3 3.5 0 2 4 6 8 10x 10 4 Calibrated Model, t =24hrs Log UI Cell Counts Data Model 0 0.5 1 1.5 2 2.5 3 3.5 0 1 2 3 4 5 6 7 8 9x 10 4 Calibrated Model, t =48hrs Log UI Cell Counts Data Model 0 0.5 1 1.5 2 2.5 3 3.5 0 0.5 1 1.5 2 2.5x 10 5 Calibrated Model, t =96hrs Log UI Cell Counts Data Model 0 0.5 1 1.5 2 2.5 3 3.5 0 0.5 1 1.5 2 2.5 3 3.5x 10 5 Calibrated Model, t =120hrs Log UI Cell Counts Data Model
(27)

Cell Numbers

0 20 40 60 80 100 120 0 2 4 6 8 10x 10

7 Total Cell Counts

Time [hrs] Number of Cells Undivided Divided Total Ni(t) = R ni(t,x) 0 20 40 60 80 100 120 0 5 10 15x 10 6 Total Precursors Time [hrs] Number of Precursors Undivided Divided Total Pi(t) =Ni(t)/2i

Population doubling time and precursor viability easily computable

(28)

Label Structure Fragmentation Model-Model 1

CFSE label dynamics can be described by the fragmentation equation ∂n(t,x) ∂tcekt∂[(x −xa)n(t,x)]x =(α(t,x) +β(t,x))n(t,x)χ[xa,x]4α(t,2xxa)n(t,2xxa) n(0,x) = Φ(x) n(t,xmax) =0 v(t,xa)n(t,xa) =0. (1)

where n(t,x)is the structured population density (cells per unit FI) at time t and measured FI x and the advection term (with parameters c and k ) represents the Gompertz decay process for decrease in FI resulting from intracellular turnover of CFSE.

(29)

Compart. Models with Generation Structure-Model 2

A model can be reformulated with distinct compartments for each generation. The resulting model is a system of partial differential equations ∂n0 ∂tcekt (xxa) ∂n0 ∂x =−(α0(t) +β0(t)−cekt )n0(t,x) ∂n1 ∂tcekt(xxa) ∂n1 ∂x =−(α1(t) +β1(t)−cekt)n 1(t,x) +R1(t,x) .. . ∂nimax ∂tcekt(xxa) ∂nimax ∂x =−(βimax(t)−cekt)n imax(t,x) +Rimax(t,x), (2)
(30)

Compartmental Models-Model 2

Here ni(t,x)is structured population density for cells having

undergone i divisions. The recruitment terms describe the symmetric division of CFSE upon mitosis. Given by Ri(t,x) =4αi−1(t)ni−1(t,2xxa). Boundary and initial

conditions same as in (1).

Observe that, because the number of divisions undergone has now been explicitly identified by the subscripted generation number, no longer necessary for division and death rates to depend upon measured fluorescence intensity.

(31)

Compartmental Models-Model 3

IDEA: Combine compartmental models with probabilistic structures (so-called Cyton Models) for cell division and death

Let Ni(t), 0iimaxrepresent the number of cells having

undergone i divisions at time t. Assume there are N0cells in

the population at t =0. In its simplest form, the cyton model

relates the number of cells in the population to the number of cells which divide and die in a unit of time,

N0(t) =N0− Z t 0 ndiv0 (s)ndie0 (s)ds Ni(t) = Z t 0

2ndivi1(s)ndivi (s)ndiei (s)ds, (3) where ndivi (t)and ndiei (t)indicate the numbers per unit time of cells having undergone i divisions that divide and die,

(32)

Compartmental Models-Model 3

Letφ0(t)andψ0(t)be probability density rate functions (in units

1/time) for the times to division and death, respectively, for an undivided cell. Let F0, the initial precursor fraction, be the

fraction of undivided cells which would hypothetically divide in the absence of any cell death. It follows that

ndiv0 (t) =F0N0 1 Z t 0 ψ0(s)ds φ0(t) ndie0 (t) =N0 1F0 Z t 0 φ0(s)ds ψ0(t). (4)

(33)

Compartmental Models-Model 3

Can define probability density rate functionsφi(t)andψi(t)for

times to division and death, respectively, for cells having undergone i divisions, as well as the progressor fractions Fi of

cells which would complete the ith division in the absence of

cell death. Then the numbers per unit time of dividing and dying cells are computed as

ndivi (t) =2Fi Z t 0 nidiv−1(s) 1 Z ts 0 ψi(ξ)dξ φi(t−s)ds ndiei (t) =2 Z t 0 ndivi1(s) 1Fi Z ts 0 φi(ξ)dξ ψi(t−s)ds. (5)

(34)

Compartmental Models-Model 4

Allg¨ower, et al., have proposed a model which is structured by the fluorescence intensity resulting from the mass of CFSE within the cell, i.e., intracellular label ignoring autofluorescence. This leads to a system for the mass of fluorescence n(t,x) described by the system of equations

n0 ∂tcekt∂[xn0] ∂x =−(α0(t) +β0(t))n0(t,x) .. . ∂nitcekt∂[xni] ∂x =−(αi(t) +βi(t))ni(t,x) +4αi−1(t)ni−1(t,2x) (6) .. .

(35)

Compartmental Models-Model 4

The major advantage of formulating the model in terms of mass of FI is the very simple form of the model solution. The solution to the model (6) can be written as

ni(t,x) =Ni(t)¯ni(t,x) (7)

for all i. In this representation the functions Ni(t)satisfy the

weakly coupled system of ordinary differential equations dN0 dt =−(α0(t) +β0(t))N0(t) dN1 dt =−(α1(t) +β1(t))N1(t) +2αi−1(t)Ni−1(t) (8) .. .

(36)

Compartmental Models-Model 4

The functionsn¯i(t,x)each satisfy the partial differential

equation

∂¯ni(t,x)

tce

kt∂[xn¯i(t,x)]

x =0 (9)

with initial condition ¯

ni(0,x) =

2iΦ(2ix) N0

.

Note that, by definition,

N0=

Z ∞

0

(37)

Compartmental Models-Model 4

Let˜ni(t,x˜)be the structured density relative to the measured

fluorescence (˜x is related to x by the addition of cellular

autofluorescence). To account for autofluorescence and because autofluorescence may vary from cell to cell in the population, this is most accurately treated by computing the convolution integral ˜ n(t,x˜) = Z ∞ 0 n(t,x)p(t,x˜x)dx, (10)

where p(t,x)˜ is (for each fixed time t) a probability density function describing the distribution of autofluorescence in the population.

(38)

Compartmental Models-Model 4

So finally we consider the system of equations

n0 ∂tcekt∂[xn0] ∂x = ndiv0 (t)ndie0 (t)n¯0(t,x) ∂n1 ∂tcekt∂[xn1] ∂x =

2n0div(t)ndiv1 (t)ndie1 (t)n¯1(t,x)

(11) ..

.

where the definitions of n0div(t), ndie0 (t), ndivi (t)and ndiei (t)are given in Equations (4) - (5).

(39)

Compartmental Models-Model 4

This model, which is based onsimple mass balance, can be

solved byfactorization(7) as before; thelabel densities n(t,x)

arereadily computedaccording to Equation (9), and thecell numbersare now provided by thecyton modelas discussed

above. Thus the new model (11) is capable ofaccurately

describing the evolving generation structureof a population of cells while also accounting for the manner in which the CFSE

profile of the population changes in time. The model iseasily

relatable to biologically meaningful parameters (times to division and death)and can besolved efficientlyso that it is eminently suitable for use in an inverse problem.

(40)

The Statistical Model

Links the mathematical model to the data

Implications for estimation procedure (Least squares vs. weighted least squares)

Nkj =I[˜n](tj,zkj;θ0) + (I[˜n](tj,zkj;θ0))γEkj

Early efforts using constant variance (CV) model, Var(Ekj) =σ02(⇒Absolute Error: Ykj =model+Ekj)

Also tried constant coefficient of variance (CCV), Var(Ekj) =σ02I[˜n](tj,zkj;θ0)2(⇒Relative

Error:Ykj =model×(1+Ekj))

Found in between error most closely described data, Var(Ekj) =σ02I[˜n](tj,zkj;θ0)(⇒SquareRoot Error:

Ykj =model+

(41)

Residual Plots

γ

=

0 vs.

γ

=

1

0 0.5 1 1.5 2 2.5 x 105 −6 −4 −2 0 2 4 6x 10 4 Residuals vs Model Model Solution Residuals 0 0.5 1 1.5 2 2.5 x 105 −10 −5 0 5 10

Modified Residuals vs Model

Model Solution

(42)

Residual Plots

0 0.5 1 1.5 2 x 104 −5000 0 5000 Residuals vs Model Model Solution Residuals 0 0.5 1 1.5 2 x 104 −40 −30 −20 −10 0 10 20 30 40

Modified (sqrt) Residuals vs Model

Model Solution

Modified (sqrt) Residuals

Nkj =I[˜n](tj,zkj;q0) + (I[˜n](tj,zkj;q0))γEkj withγ =0 (left) vs.

(43)

Least Squares Estimation

Define qWLS=arg min qQJ(q;{λj}) =arg min qQ X j,k λjI[ˆn](tj,zk;q)Nkj 2 wkj where wkj =    λjbjBˆI[ˆn](tj,zkj;q0), I[n](tj,zkj;q0)>I∗ λjˆbjBI∗, I[n](tj,z j k;q0)≤I∗ .

Then qWLSis a consistent estimator of q0(Banks, Kenz,

(44)

Fit to Data

0 1 2 3 4 5 6 0 1000 2000 3000 4000 5000

Approximated Initial Condition, t =23.5 hrs

Log Fluorescence Intensity [log UI]

Counted Cells

4σ Region Data Model

(45)

Fit to Data (cont’d)

0 1 2 3 4 5 6 0 1000 2000 3000 4000 5000 Fit to Data, t =46 hrs

Log Fluorescence Intensity [log UI]

Counted Cells

4σ Region Data Model

(46)

Fit to Data (cont’d)

0 1 2 3 4 5 6 0 1000 2000 3000 4000 5000 Fit to Data, t =67.5 hrs

Log Fluorescence Intensity [log UI]

Counted Cells

4σ Region Data Model

(47)

Fit to Data (cont’d)

0 1 2 3 4 5 6 0 1000 2000 3000 4000 5000 Fit to Data, t =94.5 hrs

Log Fluorescence Intensity [log UI]

Counted Cells

4σ Region Data Model

(48)

Fit to Data (cont’d)

0 1 2 3 4 5 6 0 1000 2000 3000 4000 5000 Fit to Data, t =117.5 hrs

Log Fluorescence Intensity [log UI]

Counted Cells

4σ Region Data Model

(49)

Fit to Data, CD8 Cells

0 1 2 3 4 5 6 0 2000 4000 6000 8000 10000 Fit to Data, t =94.5 hrs

Log Fluorescence Intensity [log UI]

Counted Cells 4σ Region Data Model 0 1 2 3 4 5 6 0 2000 4000 6000 8000 Fit to Data, t =94.5 hrs

Log Fluorescence Intensity [log UI]

Counted Cells

4σ Region Data Model

(50)

Cytons

0 20 40 60 80 100 120 0 0.005 0.01 0.015 0.02

Time to Division/Death for Undivided Cells

Time [hrs] Probability Density 0 20 40 60 80 100 120 −0.4 −0.2 0 0.2 0.4 Time to Division/Death Time [hrs] Probability Density

Donor 2, CD8 cells, undivided (left) and divided (right), Data Set 121

(51)

Generation Structure

0 20 40 60 80 100 120 0 2 4 6 8x 10 5 Time [hrs] Number of Cells Cell Numbers 0 1 2 3 4 5 6+ 0 20 40 60 80 100 120 0 2 4 6 8 10x 10 5 Time [hrs] Number of Cells Cell Numbers 0 1 2 3 4 5 6+ 0 20 40 60 80 100 120 0 1 2 3 4 5 6x 10 5 Time [hrs] Number of Cells Cell Numbers 0 1 2 3 4 5 6+ 0 20 40 60 80 100 120 0 2 4 6 8x 10 5 Time [hrs] Number of Cells Cell Numbers 0 1 2 3 4 5 6+

Donor 1 (top) and Donor 2 (bottom), CD4 cells (left) and CD8 cells (right)

(52)

Experimental Extensions

Account for multiple cell cultures present in PBMC culture Antigen-specific stimulation

Effects of cryopreservation

Extracellular signaling, knockout experiments In vitro vs in vivo differences

Linking to immune process models

(53)

Acknowledgements

National Institute of Allergy and Infectious Disease grant NIAID 9R01AI071915

U.S. Air Force Office of Scientific Research under grant AFOSR-FA9550-09-1-0226

References

Related documents