Photothermal modulation of human stem cells using light-responsive … · Photothermal modulation...

10
Photothermal modulation of human stem cells using light-responsive 2D nanomaterials James K. Carrow a,1 , Kanwar Abhay Singh a,1 , Manish K. Jaiswal a , Adelina Ramirez a , Giriraj Lokhande a , Alvin T. Yeh a , Tapasree Roy Sarkar b , Irtisha Singh a,c,2 , and Akhilesh K. Gaharwar a,d,e,2 a Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843; b Department of Biology, Texas A&M University, College Station, TX 77843; c Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, Texas A&M University, Bryan, TX 77807; d Materials Science and Engineering, College of Engineering, Texas A&M University, College Station, TX 77843; and e Center for Remote Health Technologies and Systems, Texas A&M University, College Station, TX 77843 Edited by Catherine J. Murphy, University of Illinois at UrbanaChampaign, Urbana, IL, and approved April 10, 2020 (received for review August 18, 2019) Two-dimensional (2D) molybdenum disulfide (MoS 2 ) nanomateri- als are an emerging class of biomaterials that are photoresponsive at near-infrared wavelengths (NIR). Here, we demonstrate the ability of 2D MoS 2 to modulate cellular functions of human stem cells through photothermal mechanisms. The interaction of MoS 2 and NIR stimulation of MoS 2 with human stem cells is investigated using whole-transcriptome sequencing (RNA-seq). Global gene ex- pression profile of stem cells reveals significant influence of MoS 2 and NIR stimulation of MoS 2 on integrins, cellular migration, and wound healing. The combination of MoS 2 and NIR light may pro- vide new approaches to regulate and direct these cellular func- tions for the purposes of regenerative medicine as well as cancer therapy. two-dimensional (2D) nanomaterials | whole-transcriptome sequencing | cellnanoparticles interactions | human mesenchymal stem cells | cell adhesion T wo-dimensional (2D) nanomaterials are an emerging class of materials with anisotropic layered structure that have shown distinct physical and chemical properties (14). Recently, 2D transition metal dichalcogenides (TMDs) such as molybdenum disulfide (MoS 2 ) have been explored for biomedical applications due to their photothermal characteristics (59). Specifically, the generation of ultrathin sheets via exfoliation of MoS 2 enables the formation of direct bandgap nanosheets, which can be utilized for light-based therapies (8, 1013). In addition, the absence of dangling bonds from terminal sulfur atoms of MoS 2 nano- materials render high physiological stability (6, 10). The photo- responsive ability of 2D MoS 2 along with its chemical stability in biological microenvironments can be exploited for the photo- modulation of cellular functions. Although some studies have investigated cellular compatibility of 2D MoS 2 (1417), no studies have explored their photomodulation ability. Understanding cellular responses following treatment with 2D MoS 2 , with and without photostimulation, will assist in exploring their biomedical applications. Although molecular biology tech- niques such as PCR and microarrays can be used to evaluate the effect of this treatment on cells, they are limited in throughput. To overcome this limitation, omicstechniques can provide global high-throughput readouts of various cellular and molecular pro- cesses and can help discern affected cellnanomaterials interac- tions. Specifically, whole-transcriptome sequencing (RNA-seq) of treated cells can provide an unbiased and global perspective of stimulated cellular and molecular activity as well as pivotal sig- naling pathways triggered by exposure to the nanomaterial. RNA- seq can be used for determination and quantification of all expressed transcripts in cells by overcoming the limitations and biases of conventional PCR and microarrays analysis. Here, we report interactions of exfoliated MoS 2 with human mesenchymal stem cells (hMSCs) by uncovering molecular targets and affected signaling pathways. We first examine the interplay of 2D MoS 2 with biomolecules under physiological conditions. Subsequently, we utilize RNA-seq to characterize the in-depth and unbiased effect of MoS 2 on the gene expression program of hMSCs. RNA-seq reveals the steady-state expression of the transcriptome after the MoS 2 treatment, as well as the photothermal effect of MoS 2 at near-infrared wavelengths (NIR). This information assists in identifying the role of MoS 2 and NIR exposure in influencing key signaling pathways. Biophysical Characterization of MoS 2 Nanosheets Bulk MoS 2 is composed of atomically thin SMoS sheets stacked by short-range van der Waals forces. Bulk MoS 2 was chemically exfoliated following previously reported protocols (3, 18, 19). X-ray diffraction (XRD) of bulk MoS 2 showed charac- teristic peaks at (002), (004), (103), (006), (105), and (008), while peaks broadened postexfoliation at (002), (100), and (110) as indexed by Joint Committee on Powder Diffraction File card number 37-1492 (Fig. 1A). The peaks of exfoliated MoS 2 in- dicated a hexagonal structure. Using Braggs equation [2d sin(θ) = nλ], the interlayer spacing [(002) Miller indices spacing] of exfoliated MoS 2 is 6.328 Å, which is larger than the interlayer spacing of bulk MoS 2 (6.124 Å). The full width at half maximum of peak (002) was 0.089 rad for bulk MoS 2 , which increased to 2.493 rad for exfoliated MoS 2 . The crystallite sizes for bulk and Significance We demonstrate the ability of two-dimensional transition metal dichalcogenides (TMDs) such as molybdenum disulfide (MoS 2 ) to modulate and direct cellular functions of human stem cells through photothermal modulation. The global gene ex- pression profile of stem cells reveals significant influence on integrins, cellular migration, and wound healing by photo- thermal modulation of MoS 2 . The photostimulation of MoS 2 may provide new approaches to regulate cellular migration and related functions. Author contributions: J.K.C., K.A.S., I.S., and A.K.G. designed research; J.K.C., K.A.S., M.K.J., A.R., G.L., and I.S. performed research; A.K.G. contributed new reagents/analytic tools; J.K.C., K.A.S., G.L., A.T.Y., T.R.S., I.S., and A.K.G. analyzed data; and J.K.C., K.A.S., I.S., and A.K.G. wrote the paper. The authors declare no competing interest. This article is a PNAS Direct Submission. This open access article is distributed under Creative Commons Attribution-NonCommercial- NoDerivatives License 4.0 (CC BY-NC-ND). Data deposition:The data reported in this paper have been deposited in the Gene Ex- pression Omnibus (GEO) database, https://www.ncbi.nlm.nih.gov/geo (accession no. GSE141456). 1 J.K.C. and K.A.S contributed equally to this work. 2 To whom correspondence may be addressed. Email: [email protected] or isingh@ tamu.edu. This article contains supporting information online at https://www.pnas.org/lookup/suppl/ doi:10.1073/pnas.1914345117/-/DCSupplemental. First published May 27, 2020. www.pnas.org/cgi/doi/10.1073/pnas.1914345117 PNAS | June 16, 2020 | vol. 117 | no. 24 | 1332913338 ENGINEERING Downloaded by guest on August 6, 2020

Transcript of Photothermal modulation of human stem cells using light-responsive … · Photothermal modulation...

Page 1: Photothermal modulation of human stem cells using light-responsive … · Photothermal modulation of human stem cells using light-responsive 2D nanomaterials James K. Carrowa,1 ,

Photothermal modulation of human stem cells usinglight-responsive 2D nanomaterialsJames K. Carrowa,1

, Kanwar Abhay Singha,1, Manish K. Jaiswala, Adelina Ramireza, Giriraj Lokhandea,

Alvin T. Yeha, Tapasree Roy Sarkarb, Irtisha Singha,c,2, and Akhilesh K. Gaharwara,d,e,2

aBiomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843; bDepartment of Biology, Texas A&M University, CollegeStation, TX 77843; cDepartment of Molecular and Cellular Medicine, Texas A&M Health Science Center, Texas A&M University, Bryan, TX 77807; dMaterialsScience and Engineering, College of Engineering, Texas A&M University, College Station, TX 77843; and eCenter for Remote Health Technologies andSystems, Texas A&M University, College Station, TX 77843

Edited by Catherine J. Murphy, University of Illinois at Urbana–Champaign, Urbana, IL, and approved April 10, 2020 (received for review August 18, 2019)

Two-dimensional (2D) molybdenum disulfide (MoS2) nanomateri-als are an emerging class of biomaterials that are photoresponsiveat near-infrared wavelengths (NIR). Here, we demonstrate theability of 2D MoS2 to modulate cellular functions of human stemcells through photothermal mechanisms. The interaction of MoS2and NIR stimulation of MoS2 with human stem cells is investigatedusing whole-transcriptome sequencing (RNA-seq). Global gene ex-pression profile of stem cells reveals significant influence of MoS2and NIR stimulation of MoS2 on integrins, cellular migration, andwound healing. The combination of MoS2 and NIR light may pro-vide new approaches to regulate and direct these cellular func-tions for the purposes of regenerative medicine as well ascancer therapy.

two-dimensional (2D) nanomaterials | whole-transcriptome sequencing |cell–nanoparticles interactions | human mesenchymal stem cells | celladhesion

Two-dimensional (2D) nanomaterials are an emerging class ofmaterials with anisotropic layered structure that have shown

distinct physical and chemical properties (1–4). Recently, 2Dtransition metal dichalcogenides (TMDs) such as molybdenumdisulfide (MoS2) have been explored for biomedical applicationsdue to their photothermal characteristics (5–9). Specifically, thegeneration of ultrathin sheets via exfoliation of MoS2 enables theformation of direct bandgap nanosheets, which can be utilizedfor light-based therapies (8, 10–13). In addition, the absence ofdangling bonds from terminal sulfur atoms of MoS2 nano-materials render high physiological stability (6, 10). The photo-responsive ability of 2D MoS2 along with its chemical stability inbiological microenvironments can be exploited for the photo-modulation of cellular functions. Although some studies haveinvestigated cellular compatibility of 2D MoS2 (14–17), nostudies have explored their photomodulation ability.Understanding cellular responses following treatment with 2D

MoS2, with and without photostimulation, will assist in exploringtheir biomedical applications. Although molecular biology tech-niques such as PCR and microarrays can be used to evaluate theeffect of this treatment on cells, they are limited in throughput. Toovercome this limitation, “omics” techniques can provide globalhigh-throughput readouts of various cellular and molecular pro-cesses and can help discern affected cell–nanomaterials interac-tions. Specifically, whole-transcriptome sequencing (RNA-seq) oftreated cells can provide an unbiased and global perspective ofstimulated cellular and molecular activity as well as pivotal sig-naling pathways triggered by exposure to the nanomaterial. RNA-seq can be used for determination and quantification of allexpressed transcripts in cells by overcoming the limitations andbiases of conventional PCR and microarrays analysis.Here, we report interactions of exfoliated MoS2 with human

mesenchymal stem cells (hMSCs) by uncovering molecular targetsand affected signaling pathways. We first examine the interplayof 2D MoS2 with biomolecules under physiological conditions.

Subsequently, we utilize RNA-seq to characterize the in-depth andunbiased effect of MoS2 on the gene expression program of hMSCs.RNA-seq reveals the steady-state expression of the transcriptomeafter the MoS2 treatment, as well as the photothermal effect ofMoS2 at near-infrared wavelengths (NIR). This information assistsin identifying the role of MoS2 and NIR exposure in influencing keysignaling pathways.

Biophysical Characterization of MoS2 NanosheetsBulk MoS2 is composed of atomically thin S–Mo–S sheetsstacked by short-range van der Waals forces. Bulk MoS2 waschemically exfoliated following previously reported protocols (3,18, 19). X-ray diffraction (XRD) of bulk MoS2 showed charac-teristic peaks at (002), (004), (103), (006), (105), and (008), whilepeaks broadened postexfoliation at (002), (100), and (110) asindexed by Joint Committee on Powder Diffraction File cardnumber 37-1492 (Fig. 1A). The peaks of exfoliated MoS2 in-dicated a hexagonal structure. Using Bragg’s equation [2d sin(θ)= nλ], the interlayer spacing [(002) Miller indices spacing] ofexfoliated MoS2 is 6.328 Å, which is larger than the interlayerspacing of bulk MoS2 (6.124 Å). The full width at half maximumof peak (002) was 0.089 rad for bulk MoS2, which increased to2.493 rad for exfoliated MoS2. The crystallite sizes for bulk and

Significance

We demonstrate the ability of two-dimensional transitionmetal dichalcogenides (TMDs) such as molybdenum disulfide(MoS2) to modulate and direct cellular functions of human stemcells through photothermal modulation. The global gene ex-pression profile of stem cells reveals significant influence onintegrins, cellular migration, and wound healing by photo-thermal modulation of MoS2. The photostimulation of MoS2may provide new approaches to regulate cellular migrationand related functions.

Author contributions: J.K.C., K.A.S., I.S., and A.K.G. designed research; J.K.C., K.A.S.,M.K.J., A.R., G.L., and I.S. performed research; A.K.G. contributed new reagents/analytictools; J.K.C., K.A.S., G.L., A.T.Y., T.R.S., I.S., and A.K.G. analyzed data; and J.K.C., K.A.S., I.S.,and A.K.G. wrote the paper.

The authors declare no competing interest.

This article is a PNAS Direct Submission.

This open access article is distributed under Creative Commons Attribution-NonCommercial-NoDerivatives License 4.0 (CC BY-NC-ND).

Data deposition:The data reported in this paper have been deposited in the Gene Ex-pression Omnibus (GEO) database, https://www.ncbi.nlm.nih.gov/geo (accession no.GSE141456).1J.K.C. and K.A.S contributed equally to this work.2To whom correspondence may be addressed. Email: [email protected] or [email protected].

This article contains supporting information online at https://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1914345117/-/DCSupplemental.

First published May 27, 2020.

www.pnas.org/cgi/doi/10.1073/pnas.1914345117 PNAS | June 16, 2020 | vol. 117 | no. 24 | 13329–13338

ENGINEE

RING

Dow

nloa

ded

by g

uest

on

Aug

ust 6

, 202

0

Page 2: Photothermal modulation of human stem cells using light-responsive … · Photothermal modulation of human stem cells using light-responsive 2D nanomaterials James K. Carrowa,1 ,

0 1 2 3-1

0

1

2

3

4

Distance (μm)

Hei

ght (

nm)

160 162 164

).u.a(ytisnetnI

Binding energy (eV)

BulkExfoliatedS 2p3/2

S 2p1/2

226 230 234

Inte

nsity

(a.u

.)

Binding energy (eV)

BulkExfoliated

Ram

an In

tens

ity (a

.u.)

200 300 400 500

BulkExfoliated

J3

E1g

J2

E12g

A1g

Control (no MoS ) MoS (2 mg/mL)

Ther

mal

Imag

ing

15 mm

Time (min)

Tem

pera

ture

(°C

)

0 1 2 3 4 5 6 7 8 9 10 11

22

24

26

28 Control (water)

Control (media)2 mg/mL MoS2 (340 mW/cm2)2 mg/mL MoS2 (20 mW/cm2)

500 g/mL MoS2 (340 mW/cm2)

500 g/mL MoS2 (20 mW/cm2)

MoS (500 g/mL)

PL

Inte

nsity

(a.u

.)

400 450 500 550 6000

10000

20000

30000

40000 Protein + SDS (-control)Protein only (+control)Protein + 50 g/ml MoS2

Protein + 100 g/ml MoS2

Protein + 150 g/ml MoS2

MoS2 only

Wavelength (nm)400 450 500 550 600

Wavelength (nm)Water

PBSMed

ia0

200

400

600

800

Hyd

rody

nam

ic d

iam

eter

(nm

)

Water

PBSMed

ia-40

-30

-20

-10

0

Zeta

Pot

entia

l (m

V)

Sample Mo S

Bulk 14.8 % 85.2 %

Exfoliated 22.8 % 77.2 %

Exfoliated Molybdenum Disulfide (MoS2)

Atomic ( %)

Bulk Molybdenum Disulfide (MoS2)

10 20 30 40 50 60 702 (degrees)

(002

)

(100

)

(110

)

Exfoliated MoS2

Inte

nsity

(a.u

)

10 20 30 40 50 60 702 (degrees)

(002

)

(004

)

(103

)(0

06)

(105

)

(008

)

Bulk MoS2

Inte

nsity

(a.u

)

1 m5 m

1

2

3

100 nm

100

103

105

110200

10 nm-1

Raman Shift (cm-1)600 650 700 750 800

BulkExfoliated

Wavelengh (nm)

).u.a(ytisnetnI

dezilamro

N

Mo 3d5/2

Mo 3d

Photo-thermal stimulation via near infrared (NIR) (λ = 800 nm, P = 340 mW/cm2, t = 7 mins)

A

B

F

C

D

H

I J

GE

*

*

* *

Fig. 1. Physicochemical characterization of exfoliate 2D MoS2 nanosheets. (A) XRD of bulk and exfoliated MoS2. (B) AFM measures micrometer-sizednanosheets with nanometer thickness, confirming 2D shape. (C) TEM images of ultrathin MoS2 sheets displayed overlapping layers; each individual layeris numbered. Further, electron diffraction corroborated the formation of the 1T phase following lithium intercalation. (D) Atomic composition of bulk andexfoliated MoS2 determined via elemental analysis. (E) Crystallographic transformations from bulk to exfoliated MoS2 were monitored with XPS analysis withshifts from 2H to 1T phases observed. (F) Raman spectroscopy and (G) photoluminescent measurements likewise indicated structural modifications hadoccurred, seen through changes in vibrational energy bands and luminescent intensity, respectively. (H) Response to NIR light was determined using an IRcamera and change in temperature over time. (I) Zeta potential and hydrodynamic size of exfoliated MoS2 in water, PBS, and media. (J) ANS assay showsprotein structures are intact in the presence of exfoliated MoS2.

13330 | www.pnas.org/cgi/doi/10.1073/pnas.1914345117 Carrow et al.

Dow

nloa

ded

by g

uest

on

Aug

ust 6

, 202

0

Page 3: Photothermal modulation of human stem cells using light-responsive … · Photothermal modulation of human stem cells using light-responsive 2D nanomaterials James K. Carrowa,1 ,

exfoliated MoS2 were calculated using Scherrer equation as∼120 nm and 3.5 nm, respectively.The morphology of MoS2 nanosheets was characterized by

atomic force microscopy (AFM) and transmission electron mi-croscopy (TEM). AFM measured layered sheets that were 2 to7 μm in diameter and ∼3 nm thick (Fig. 1B). The theoreticalcalculation suggested the S–Mo–S atomic lattice thickness to beabout 6.5 Å (8), which indicated the selected nanoflake in themagnified AFM image contained three to five sheets of mono-layer MoS2. TEM images indicated smooth topography andcrystalline structure of MoS2 (Fig. 1C). The selected area elec-tron diffraction (SAED) pattern of monolayer MoS2 confirmedthe formation of 1T phase after exfoliation. Characteristic SAEDpeaks agreed with XRD data indicating a hexagonal structure.The elemental composition of bulk (14.8% Mo and 85.2% S)

and exfoliated MoS2 (22.8% Mo and 77.8% S) was determinedby X-ray photoelectron spectra (XPS) (Fig. 1D). The bindingenergies (BE) for molybdenum and sulfur core-level electronsrevealed the crystallographic phases associated with bulk MoS2transformed to a few layers (Fig. 1E). The BE for Mo 3d dou-blets (3d5/2 and 3d3/2) were at 228.5 and 231.7 eV for bulk MoS2,which, after exfoliation, shifted to 228.2 and 231.5 eV, re-spectively, indicating the 2H to 1T phase transformation. Simi-larly, the BE for S 2p doublets (2p3/2 and 2p1/2) were 161.4 and162.6 eV, which correlate with the 2H phase and shifted to 161.1and 162.3 eV, respectively, following exfoliation indicating1T phase.Raman spectroscopy confirmed the vibrational modes associ-

ated with the MoS2 lattices for both bulk and exfoliated sheets(Fig. 1F). As expected, the two characteristic modes, E1

2g cor-responding to in-plane vibrations of sulfur atoms with respect tomolybdenum and A1g corresponding to out-of-plane vibration ofsulfur atoms, had energies of 380.8 and 406.5 cm−1, respectively,in bulk form. Upon exfoliation, the E1

2g shifted to 376.5 cm−1

and the A1g band shifted to 403.9 cm−1. The relative magnitudeof out-of-plane (A1g) vibration was larger than in-plane vibration(E1

2g) in bulk. After layer exfoliation, the relative magnitude ofin-plane vibration (E1

2g) was larger than out-of-plane vibration(A1g), corroborating more free sulfur atoms. Furthermore, due tothe phase change from 2H to 1T, additional vibrational bandssuch as E1g, J2, and J3 were also observed for the exfoliated MoS2sample (20).Furthermore, photoluminescent intensity decreased for exfo-

liated MoS2 samples due to Li intercalation, resulting in a phasetransition from semiconducting 2H to metallic 1T phase(Fig. 1G). This corroborates the observation of phase transitionfrom 2H to 1T in the Raman spectra. The NIR responsivenesswas evaluated to ensure photothermal effects could be utilizedfor cell stimulation. A solution of MoS2 (500 μg/mL and 2 mg/mL) was subjected to NIR light, and the solution temperaturewas monitored using a thermocouple and IR imaging. After10 min of exposure to NIR light, the temperature of MoS2 so-lution (2 mg/mL) was increased by ∼5.5 °C and ∼1.5 °C for thehigh- (340 mW/cm2) and low- (20 mW/cm2) power NIR light,respectively (Fig. 1H). For low concentration of MoS2 (500 μL/mL)and water, no significant change in temperature was observed. Thisdemonstrated that photothermal effects could be expected withina local cellular environment following NIR exposure to exfoliatedMoS2 nanosheets.

Physicochemical Stability of MoS2 NanosheetsThe aqueous stability of exfoliated MoS2 nanosheets was eval-uated in deionized water (diH2O), phosphate-buffered saline(PBS), and cell culture media containing fetal bovine serum.MoS2 nanosheets were stable in diH2O and PBS and showedstrong negative zeta-potential measurements of −32.7 ± 1.1 mVand −29.2 ± 1.7 mV for diH2O and PBS, respectively (Fig. 1I).After the addition of protein, however, the surface of shear, or

the interface of bound charges and diffuse layer, is shifted furtherfrom the surface, effectively reducing the zeta potential. Thisphenomenon was observed as the zeta potential significantlydropped to −17.3 ± 0.8 mV after subjecting MoS2 nanosheets tocell culture media. This indicated that while biomolecules andmultivalent ions were strongly interacting with the nanomaterialsurface, the repulsive forces between MoS2 nanosheets are reducedvia charge screening, leading to eventual flocculation at longertimescales.To improve nanosheet dispersions within these aqueous media

environments, bath or probe sonication can be utilized. Thesemechanical methods to reduce interactions between sheets mayalso reduce the effective sheet size. To investigate the effects oftype and duration of sonication, dynamic light scattering (DLS)was utilized on aqueous dispersions of MoS2. Samples lackingsonication displayed relatively large nanosheet sizes with an av-erage hydrodynamic diameter of 6.9 ± 0.6 μm. Following soni-cation (2 min), there was a significant reduction in size to 2.6 ±0.8 μm (bath sonication) and 0.6 ± 0.03 μm (probe sonication).Extending sonication (10 min) further reduced the hydrody-namic size to 1.0 ± 0.3 μm (bath sonication) and 0.38 ± 0.02 μm(probe sonication). Significant reduction in physical size within10 min of sonication has important ramifications for samplepreparation prior to cellular exposure. Specifically, even a briefsonication to improve dispersions of the nanosheets may result inphysical changes to the MoS2. Smaller MoS2 nanosheet sizes willaffect binding dynamics to proteins in the aqueous environmentand subsequently to the cell membrane. Furthermore, reductionin hydrodynamic size as a result of sonication facilitates im-proved uptake of the nanosheets. Hence, we selected probesonication for 10 min for MoS2 (∼400-nm-long and 3- to4-nm-thick nanosheet) for all future studies.As previously noted, the hydrodynamic size of MoS2 was

influenced by the presence of ions (PBS and media) and protein(media). The adsorption of protein on the MoS2 nanosheet willinfluence cellular adhesion and internalization due to formationof protein corona on the nanosheet. To evaluate these changes inprotein conformation, an 8-anilinonapthalene-1-sulfonic acid(ANS) assay (21) was utilized (Fig. 1J). ANS binds selectivecationic amino acid residues (such as arginine and lysine) viaformation of an ion pair between the positively charged residuesand the sulphonyl groups of the ANS. The formation of these ionpairs results in increased fluorescence emission, and thuschanges in conformation can be detected by measuring the rel-ative fluorescence intensity. When a model protein with positivecharge at buffered pH, lysozyme (isoelectric point –11.35) issubjected to sodium dodecyl sulfate (SDS), a known disruptor ofprotein structure, a significant change in protein conformationwas observed, as indicated by an increase in fluorescence signal.However, subjecting protein to MoS2 nanosheets resulted in aminimal change in fluorescence signal and therefore proteinconformation. Furthermore, following an increase in the con-centration of MoS2 nanosheet from 50 to 150 μg/mL, no signif-icant impact on fluorescent signal was observed, indicatingmaintenance of native protein structure over a range of relevantconcentrations. The sequestering proteins with intact nativestructure would enable clustering of receptors on the cellmembrane and stimulate downstream intracellular pathways(22, 23).

Cellular Interaction of MoS2 NanosheetsAs soon as MoS2 is subjected to physiological fluids, proteincorona formed on its surface due to high surface area and chargecharacteristics. We used gel electrophoresis to observed forma-tion of protein corona. Most of serum protein was adsorbed onMoS2 and no significant difference in protein adsorption wasobserved due to NIR treatment of MoS2. The colloidal stabilityof MoS2 in physiological conditions will enable significant

Carrow et al. PNAS | June 16, 2020 | vol. 117 | no. 24 | 13331

ENGINEE

RING

Dow

nloa

ded

by g

uest

on

Aug

ust 6

, 202

0

Page 4: Photothermal modulation of human stem cells using light-responsive … · Photothermal modulation of human stem cells using light-responsive 2D nanomaterials James K. Carrowa,1 ,

interactions of MoS2 nanosheets with the cell membrane. It isexpected that due to 2D characteristics, MoS2 can become in-ternalized by cells (Fig. 2A). We investigated cellular compati-bility of MoS2 by determining the half inhibitory concentration(IC50) using metabolic activity assay. MoS2 nanosheets showedhigh cytocompatibility at lower concentrations (<100 μg/mL)after 7 d of culture (Fig. 2B). The IC50 of MoS2 was observed tobe ∼400 μg/mL.Bright-field imaging indicated substantial binding of MoS2

nanosheets (100 μg/mL) to cell surfaces (Fig. 2C). Electron mi-croscopy was used to evaluate the extent of membrane coating by

these nanomaterials following introduction into the media. In-terestingly, scanning electron microscopy (SEM) images of thecell body depicted homogeneous coverage of submicron-sizedmaterial across the membrane. The uniform coverage of MoS2on the cell surface is expected to improve the likelihood ofphotothermal activation of cells. However, a slight decrease incell spreading was observed due to MoS2 exposure (100 μg/mL)over a period of 72 h (SI Appendix, Fig. S1).To determine the uptake mechanism of MoS2 nanosheet, an in-

hibition assay was performed using endocytosis pathway-specific in-hibitors (chlorpromazine hydrochloride to inhibit clathrin-mediated,

A

F

Bright field image SEM images

0

20

40

60

80

100

Cel

l Pop

ulat

ion

(%)

G1

S

G2

Plasma protein

Nucleus

MoS2 nanosheet (Adhesion to cell membrane and internalization via macropinocytosis)

G H

0

25

50

75

100

Cel

lPop

ulat

ion(

%)

Live

Apoptotic

Dead

Cytosol

MoS2 with protein coronaPlasma

membrane protrusion

Macropinocytosis

B

1010

25

50

75

100

MoS2 concentration ( g/mL)

Cel

l via

bility

(%

)

102 103

IC50~ 400 g/mL

E

Days of Culture

Red

uctio

nof

Ala

ma r

B lue

(Fr a

ctio

n)

1 3 70.0

0.2

0.4

0.6

0.8

0MoS2 (µg/mL)

10501005001000

**********

*****

0

20

40

60

80

100

120

Nor

mal

ised

Upt

ake

(%)

No Inhibitor

Inhibitor of clathrin-mediated uptakeInhibitor of calveolar-mediated uptake

Inhibitor of macropinocytosis

***

***

MoS2(25 g/mL) MoS2_NIR

D

0

20

40

60

80

100

120

Live

cells

(%)

n.s *

C

hMSCs

hMSCs_

Mo(50%

)

hMSCs_

Mo(100

%)

hMSCs

hMSCs_

Mo(50%

)

hMSCs_

Mo(100

%)0.0

0.5

1.0

1.5

Cel

l via

bilit

y (fr

actio

n)

**

24 hours3 hours 72 hours

MoS

2 (10

0 g/

mL)

Con

trol

hMSCs

hMSCs_

MoS 2

hMSCs

hMSCs_

MoS 2

hMSCs_

NIR

hMSCs_

NIR_M

oS 2

hMSCs

hMSCs_

MoS 2

hMSCs_

NIR

hMSCs_

MoS 2_N

IR

hMSCs_

MoS 2

hMSCs_

MoS 2

200 m

300 µm 50 µm 10 µm

Fig. 2. Cellular interactions of 2D MoS2 with hMSCs. (A) A protein corona forms when 2D MoS2 is suspended in media and adheres to the cell surface and issubsequently internalized by cells. (B) IC50 of MoS2 determined using metabolic assays. (C) Cellular internalization of MoS2 determined using optical, electron,and fluorescence microscopy. (D) MoS2 adhere to cell membrane and internalize via micropinocytosis and clathrin-mediated endocytosis. (E) Effect of MoS2 onmetabolic activity over 7 d. (F) Effect of Mo and MoS2 nanosheet at equivalent molarities on cell viability (day 3 and day 7 on the left and right, respectively).(G) Effect of MoS2 on cell cycle. (H) Cell viability when treated with NIR, MoS2, and MoS2_NIR. *P < 0.05; not significant (n.s.) > 0.05.

13332 | www.pnas.org/cgi/doi/10.1073/pnas.1914345117 Carrow et al.

Dow

nloa

ded

by g

uest

on

Aug

ust 6

, 202

0

Page 5: Photothermal modulation of human stem cells using light-responsive … · Photothermal modulation of human stem cells using light-responsive 2D nanomaterials James K. Carrowa,1 ,

nystatin to inhibit caveolar-mediated, and wortmannin to inhibitmacropinocytosis) (Fig. 2D). The results indicated that the pri-mary mechanisms of nanosheets uptake were clathrin (16 ± 3%inhibition, P < 0.05, one-way ANOVA) and macropinocytosis

(26 ± 4% inhibition, P < 0.01, one-way ANOVA). No significantdifference was observed using caveolar inhibitor as 4 ± 2% in-hibition was observed. It is also important to note that adheredMoS2 nanosheets may increase side scattering, the metric utilized

hMS

Cs_

2

hMS

Cs_

MoS

2_2

hMS

Cs_

NIR

_2

hMS

Cs_

MoS

2_N

IR_2

hMS

Cs_

1

hMS

Cs_

MoS

2_1

hMS

Cs_

NIR

_1

hMS

Cs_

MoS

2_N

IR_1

Differentially expressed genes (DEG)

MoS2(23 genes)

NIR(2 gene)

MoS2_NIR(77 genes)

MoS2 and MoS2_NIR(80 genes)

Venn diagram showing overlaping genes (FDR-adjusted P < 0.05)

−2

−1

0

1

2

0 5 10 15

−6−4

−20

24

6

Log2 (Mean of normalized counts)

Total (10996)High (27)Low (76)

0 5 10 15

−6−4

−20

24

6

Log2 (Mean of normalized counts)

Total (10996)High (1)Low (1)

0 5 10 15

−6−4

−20

24

6

Log2 (Mean of normalized counts)

Log 2F

oldC

hang

e

Total (10996)High (41)Low (116)

hMSCs_MoS2_NIR vs hMSCs

hMSCs_NIR vs hMSCs hMSCs_MoS2 vs hMSCs

Log 2F

oldC

hang

e

Log 2F

oldC

hang

e

hMSCs_MoS2 (25 µg/mL)

hMSCs_NIR

hMSCs_MoS2_NIR

hMSCs (control)

NIR

MoS2

0

5

10

15

0 5 10 15

Log 2(h

MS

Cs_

2 fp

km) r: 0.98

Log2(hMSCs_1 fpkm)

0

5

10

15

0 5 10 15Log 2(h

MS

Cs_

MoS

2_2

fpkm

)

Log2(hMSCs_MoS2_1 fpkm)

0

5

10

15

0 5 10 15

Log2(hMSCs_NIR_1 fpkm)

0

5

10

15

0 5 10 15Log 2(h

MS

Cs_

NIR

_2 fp

km)

Log2(hMSCs_MoS2_NIR_1 fpkm)

Log 2(h

MS

Cs_

MoS

2_N

IR_2

fpkm

)

r: 0.99

r: 0.99

r: 0.98

CA

B

MoS2+NIR

Row

-scaled z-score

Fig. 3. Understanding the effect of NIR, MoS2, and MoS2_NIR on hMSCs using RNA-seq. (A) Pearson correlation coefficient of expressed genes betweenreplicates of different conditions (r = 0.98 for hMSC controls, r = 0.99 for hMSCs_MoS2, r = 0.99 for hMSCs_NIR, and r = 0.98 for hMSCs_MoS2_NIR). (B) MA plothighlighting differentially regulated genes (FDR-adjusted P < 0.05) of the treatment groups against the untreated hMSCs and extent of expression change(gray: all of the expressed genes, red: up-regulated genes, blue: down-regulated genes). Venn diagram indicating differential genes overlap betweenpairwise comparison of treatment groups. (C) Heat map of row normalized FPKM z-scores of differentially expressed genes across all pairwise comparisonsagainst the untreated hMSCs (red: up-regulated, blue: down-regulated).

Carrow et al. PNAS | June 16, 2020 | vol. 117 | no. 24 | 13333

ENGINEE

RING

Dow

nloa

ded

by g

uest

on

Aug

ust 6

, 202

0

Page 6: Photothermal modulation of human stem cells using light-responsive … · Photothermal modulation of human stem cells using light-responsive 2D nanomaterials James K. Carrowa,1 ,

to monitor uptake, which may inflate uptake values and result inrelatively modest inhibition using pathway-specific inhibitors.Long-term studies indicate that hMSCs readily proliferated overa course of 7 d in the presence of various concentration of MoS2,indicating cytocompatibility even at higher concentrations (>IC50)(Fig. 2E).Earlier studies have mentioned that MoS2 degrades in physi-

ological conditions particularly in the presence of peroxidases(24). The effect of Molybdenum (Mo) was evaluated on cell vi-ability by directly exposing 50 and 100% molar concentration ofMo to cells (Fig. 2F). A significant reduction in cell viability wasobserved due to Mo exposure, while a molar equivalent of MoS2did not impact cell viability, indicating that Mo is not releasedfrom MoS2 in physiological conditions. Other indicators of cel-lular viability such as cell cycle showed no change as result ofMoS2 exposure (Fig. 2G). These results were used to determinethe concentration of MoS2 nanosheets (25 to 100 μg/mL) forsubsequent photothermal activation.To further establish the cytocompatibility of MoS2, cellular

apoptosis after exposure to MoS2 was measured using Annexin Vassay. The cells were treated with MoS2 nanosheets (25 μg/mL)for a period of 24 h with and without NIR exposure. The dataindicated that there was no significant difference in live cellpopulations between control and treated cells, irrespective ofNIR treatment (Fig. 2H). The increase in NIR intensity (340mW/cm2) can be used for photothermal therapy, which will beuseful for cancer treatment. Specifically, cells in the selectedregion can be remotely ablated by focusing NIR light (SI Ap-pendix, Fig. S2). However, here we have used low-intensity NIR(20 mW/cm2) exposure for photothermal modulation.

Global Transcriptomic Profile of Stem Cells Treated withMoS2 NanosheetsThe changes in the steady-state expression levels of cellulartranscriptome after treatment with external agents such asnanoparticles and radiation can be determined using RNA-seq(25). To investigate the effect of NIR (20 mW/cm2), MoS2 (25μg/mL), and NIR stimulation of MoS2 (MoS2_NIR), hMSCswere cultured under four different conditions including un-treated hMSCs as control (Fig. 3A). A lower concentration ofMoS2 (25 μg/mL) was selected to avoid molecular perturbationdue to cellular stress. After 7 d of treatment, the expressedmessenger RNA (mRNA) of treated cells were collected andsequenced (Materials and Methods and SI Appendix, Materialsand Methods). This early-stage time point was utilized to uncoverinitial molecular changes, as well as to capture a snapshot ofcell–nanomaterial interactions as MoS2 remained on the cellsurface up until 7 d.Using RNA-seq aligner (26), the sequenced reads were

aligned to the reference genome (hg38). The gene expressionlevels for every gene in every sample was normalized to obtainfragments per kilobase of transcript per million reads (FPKM).The correlation of expressed gene levels (FPKM) for replicatesamples subjected to different conditions indicated a high degreeof consistency (r = 0.98 for hMSC controls, r = 0.99 forhMSCs_MoS2, r = 0.99 for hMSCs_NIR, and r = 0.98 forhMSCs_MoS2_NIR). The read count for gene expression wasmodeled by generalized linear models (GLMs) (27), revealingsignificant gene expression changes between different treatmentconditions compared to untreated hMSCs (Fig. 3B and DatasetS1). NIR exposure to hMSCs (hMSCs_NIR) displayed minimaleffect on the gene expression program, as only two differentiallyexpressed genes (DEGs, false discovery rate [FDR]-adjusted P <0.05) were observed. Meanwhile application of MoS2 andMoS2_NIR resulted in more robust changes in the gene ex-pression program of hMSCs. Specifically, hMSCs treated withMoS2 and MoS2_NIR showed significant (FDR-adjusted P <0.05) change in their gene expression profile, 103 and 157 genes,

respectively. Interestingly, our previous work investigating 2Dnanomaterial treatment of hMSCs had demonstrated a highernumber of DEGs (28), indicating that the chemical and physicalcharacteristics of MoS2 affects a more selective group of cellularpathways. The DEGs in hMSCs_MoS2 and hMSCs_MoS2_NIRshowed a decent overlap (80 genes).The hierarchical clustering of DEGs highlights the concor-

dance of effect of MoS2 and MoS2_NIR treatments compared tountreated control (hMSCs) and hMSCs_NIR (Fig. 3C) acrosssamples. Overall, our results indicated that the effect of MoS2 onhMSCs is relatively moderate compared to previously reported2D nanomaterials (28). We suspect that the relatively large sizeand chemical stability of MoS2 nanomaterials limited the cell–nanomaterials interaction. However, our study suggests thatNIR-stimulated MoS2 nanomaterials may be used to regulategene expression on demand.

Photothermal Modulation of Stem Cell Gene ExpressionIn order to determine the pathways significantly impacted byMoS2 and MoS2_NIR exposure, we utilized a variety of gene setenrichment techniques including GOStats (29) and Cytoscape(30, 31). Gene ontology (GO) enrichment analysis identifiescellular functions, processes, and components that show en-richment for DEGs affected by nanomaterials and NIR treat-ment. While MoS2 treatment alone showed enrichment for 106GO terms (P < 0.05), MoS2_NIR treatment resulted in 218 GOterms (P < 0.05). Focusing largely on GO terms with high sig-nificance (P value < 0.05), we encountered key terms specific toextracellular matrix organization (GO:0030198) as well as an-giogenesis (GO:0001525) and wound healing (GO:0042060)(Fig. 4A). REVIGO analysis (32), a data management techniqueto reduce redundancies among terms from clustered genes, re-fined enrichments within the MoS2/NIR treatment group tomacromolecule metabolism, extracellular matrix (ECM) orga-nization, response to stimulus, wound healing, and regulation ofmotility and proliferation (SI Appendix, Fig. S3). These reducedterms correlated well with our GO terms of high significance anddemonstrated a clarified progression of cellular events followingnanomaterial treatment and NIR stimulation.We likewise created a gene network from coexpression data

to examine clusters of these differentially expressed genes(FDR-adjusted P < 0.05) using Cytoscape (30, 31). These clus-ters grouped genes based on known pathways and signalingfunction, thereby generating a visualization of genetic interde-pendencies among multiple cellular processes (Fig. 4B). Out ofthe pool of genes identified from differential analysis, eight clearclusters of cell function and behavior emerged pertaining toangiogenesis, vascular development, cell–cell interactions, ionhomeostasis, stress signaling, ECM organization, nucleic acidhomeostasis, and protein binding/intracellular signaling. Specif-ically, for MoS2-treated hMSCs, there were clear dependenciesbetween genes expressed within angiogenic/vascular develop-ment clusters with that of the extracellular matrix.To evaluate the activation of stress related pathways such

mitogen-activated protein kinase cascade due to NIR, MoS2, andMoS2_NIR treatment, we evaluated production of phospho-MEK1/2. MEK1/2 are dual-specificity protein kinases that me-diate the phosphorylation of tyrosine and then threonine inERK1 or ERK2. As no significant production of pMEK1/2 wasobserved, treatment with NIR, MoS2, and MoS2_NIR did notresult in activation of cellular stress-related signaling pathways(SI Appendix, Fig. S4). We also evaluated the differentiationability of hMSCs after NIR, MoS2, and MoS2_NIR treatment;however, no significant change in expression of early osteogenicmarker such as alkaline phosphate was observed (SI Appendix,Fig. S5). Overall, these results indicate that NIR, MoS2, andMoS2_NIR treatment do not cause any cellular stress.

13334 | www.pnas.org/cgi/doi/10.1073/pnas.1914345117 Carrow et al.

Dow

nloa

ded

by g

uest

on

Aug

ust 6

, 202

0

Page 7: Photothermal modulation of human stem cells using light-responsive … · Photothermal modulation of human stem cells using light-responsive 2D nanomaterials James K. Carrowa,1 ,

D

Matrix Interactions

0 2 4 6Cell-substrate adherens junction (GO:0007045)

Substrate-dependent cell migration (GO:0006929)Adherens junction assembly (GO:0034333)

Cell adhesion mediated by integrin (GO:0033627)Integrin-mediated signaling (GO:0007229)

Cell junction organization (GO:0034330)Cell junction assembly (GO:0034329)

Cell-substrate junction assembly (GO:0007044)Cell-substrate adhesion (GO:0031589)

Cell-matrix adhesion (GO:0007160)

-log10 (p-value)

Angiogenesis

0 2 4 6 8 10Regulation of blood vessel size (GO:0050880)

Endothelial cell migration (GO:0043534)Regulation of blood vessel (GO:0097746)

Sprouting angiogenesis (GO:0002040)Blood vessel morphogenesis (GO:0048514)

Angiogenesis (GO:0001525)Cardiovascular system development (GO:0072358)

Vasculature development (GO:0001944)Blood vessel development (GO:0001568)

Response to wounding (GO:0009611)Wound healing (GO:0042060)

-log10 (p-value)

Vasculature Development

0 5 10 15Muscle tissue development (GO:0014706)

Muscle cell development (GO:0055002)Actomyosin structure organization (GO:0031032)

Smooth muscle cell prolfieration (GO:0048659)Myofibril assembly (GO:0030239)

Muscle structure development (GO:0061061)Muscle contraction (GO:0006936)

Muscle cell differntiation (GO:0042692)Supramolecular fiber organization (GO:0097435)

Muscle system process (GO:0003012)Muscle cell differentiation (GO:0051146)

Collagen fibril organization (GO:0030199)Collagen metabolic process (GO:0032963)Collagen catabolic process (GO:0030574)

-log10 (p-value)

A BhMSCs_MoS2_NIR vs hMSCs

ITGA7COL5A3

COL11A1

COL4A1

ITGA11

DPYSL4COL12A1

COL16A1

COL1A1

HAPLN1

TUFT1

FBN1

LIMS2

DAG1FN1

APLP1

AGRN

COL5A2

AKR1B10

NREPPCSK7

LIMCH1

AKR1C1 NQO1

TMEM171

TP53I11

TPM1

HMGA1

IFI44

RFX8

CTPS1

CSF2RB

TSLP

IFIT1

C1orf198

LPXN

PVR

CFB TINAGL1

HAPLN3SPARC

NID2GREM1TNS1

CD248

COL5A1COL4A2

CLIC3ANXA10BDKRB1

AKR1C3

TNFSF4 EFHD1

TENM2

MASP1

ABCA3

CPNE7

SLC2A1

IDH2ITPR3

SCD5

FTH1

PXDNHMOX1

CTSC

MEGF6

CTSK ACANSPP1

FNDC1COL8A2

TFPI2

COL1A2

LAMA5ELN

IFFO2

KRT7ALS2CL

KRT14PODN

PLTP JUPLXN CDKN2B

NXPH3SCG5RARRES1

SAA1

ADAMTS7

HSPB7

PDGFRB

NPPB

SIRPA

TEK

THBS1

L1CAM

PLA2G4A

ESM1

RCAN1

MYH9

CCL2

IL8

DCBLD2

PCDH10PFKFB4

PPME1FSTL1

OSBPL10IGFBP5

STK38L

CRYAB

SCUBE3SORT1

EXTL1FHL1

LMCD1

MYH10

CKB

MGST1

LBH

GPC4

MLPHIGSF23

LRRC17PPP1R14A

LDB3

SERPINB2

STEAP1

FLG

PHLDA1 DACT1

KISS1

BST1

MAP3K7CL

FZD7DUSP6

ALDH1B1

GPNMB

ACTA2

DMPK

DKK1TPM2 SYNPO2

LMOD1

ACTG2

CNN1

Cell-CellInteractions Angiogenesis

Protein Binding/Intracellular Events

Nucelic AcidHomeostasis

Extracellular MatrixOrganization

IonHomeostasis

Stress Signaling

Vascular Development

hMSCs_MoS2_NIR vs hMSCs

0 hour 24 hours 48 hours12 hours

hMSCs

hMSCs_MoS2

hMSCs_NIR

hMSCs_MoS2_NIR

C

hMSCs hSMCs MCF-70

20

40

60

80

100 Cells

Cells_MoS2

Cells_NIR

Cells_MoS2_NIR

Wou

nd H

ealin

g C

over

age

(%)

48 hoursE

Inva

sion

Dep

th(

m)

HUVECs

HUVECs_MoS2

HUVECs_MoS2_NIR30

60

90

120

150

*****

Inva

sion

HUVECs

HUVECs_MoS 2

HUVECs_MoS 2

_NIR

CCL2COL1A1

COL1A2

COL5A1

DAG1

DCBLD2

FN1FZD7

HMOX1

ITPR3

LBH

MCAMMYH9

PDGFRBSAA1

SERPINB2

SPARC

TFPI2

THBS1TPM1

0

1

2

3

4

5

−2 0 2

Wound healing (GO:0042060)

Log2(hMSCs_MoS2_NIR/hMSCs)

-Log

10(p

-adj

ust)

100 m

100 m

Fig. 4. Functional effect of NIR, MoS2, and MoS2_NIR on cells. (A) GO terms obtained by gene enrichment analysis of DEGs (FDR-adjusted P < 0.05) due toMoS2_NIR treatment. (B) GeneMANIA analysis produced a network of coexpression interactions for differentially expressed genes within the hMSCs_MoS2_NIRpopulation (red: up-regulated, blue: down-regulated; size increases with significance). (C) A volcano plot for the wound healing (GO:0042060), gray: all of theexpressed genes, blue: genes associated with the GO term with no significant change in expression, red: genes associated with the GO term that show significantlydifferential expression due to treatment. (D) Scratch assay used to determine the effect of NIR, MoS2, and MoS2_NIR on migration, cell proliferation, and woundhealing of hMSCs, hSMCs, andMCF-7. The dotted lines represent scratch generated using pipette tip. The cell migration across the wound area was determined byimaging the sample after 12, 24, and 48 h. (E) Three-dimensional invasion assay demonstrating ability of HUVECs treated with MoS2 and MoS2_NIR to invadecollagen gel.

Carrow et al. PNAS | June 16, 2020 | vol. 117 | no. 24 | 13335

ENGINEE

RING

Dow

nloa

ded

by g

uest

on

Aug

ust 6

, 202

0

Page 8: Photothermal modulation of human stem cells using light-responsive … · Photothermal modulation of human stem cells using light-responsive 2D nanomaterials James K. Carrowa,1 ,

MoS2 and NIR Exposure Suppresses Cell Migration byModulating Cellular AdhesionBy evaluating the genes comprising highly significant GO terms,we may identify genes responsible for observable changes in cellbehavior. Among the cluster of genes related to wound healing(GO:0042060), we noted the majority of significantly DEGsbetween control cells and those receiving MoS2_NIR were down-regulated (14 out of 20) (Fig. 4C). Among these, the limb budand heart development gene (LBH) was down-regulated aftercells were introduced to MoS2 and MoS2_NIR (log2fold: −1.799and −2.846) (SI Appendix, Fig. S6). LBH plays a prominent rolein cardiogenesis as a transcriptional activator. In addition, LBHinteracts with a multitude of genes implicated in a variety ofprocesses, including vascular development and transcriptionalevents inside the nucleus.Due to this significant shift in signaling and impact on cell

machinery, we investigated the ability of cells to migrate in asimulated wound-healing experiment. Compared to controlpopulations or those receiving NIR exposure alone, hMSCs ex-posed to MoS2 and MoS2_NIR migrated into a simulated woundat significantly reduced rates (Fig. 4D and SI Appendix, Fig. S7).We further validated this effect on two different cells, humansmooth muscle cells (hSMCs) and Michigan Cancer Foundation-7 (MCF-7) breast cancer cells, with both types displaying sig-nificantly decreased migration following MoS2 and MoS2_NIRtreatment. This indicated that the reduction in cell migration dueto MoS2 and MoS2_NIR was not cell-type-specific and utilizeduniversal cell machinery that is affected by MoS2. To furtherstrengthen this hypothesis, we performed three-dimensional in-vasion assay to determine the effect of MoS2 nanosheets andNIR stimulation on human umbilical vein endothelial cells(HUVECs) migration within a collagen matrix (33). HUVECstreated with MoS2 and MoS2_NIR showed significant decreasein invasion ability as determined by the invasion depth as com-pared to control (Fig. 4E).All these experimental results validated our observations from

RNA-seq data. Specifically, GO term enrichment analysis in-dicated highly related terms, including regulation of cell motility(GO:2000145) and negative regulation of locomotion (GO:0040013).Within these GO terms for the MoS2_NIR group, genes likePODN (log2fold: −1.526), GREM1 (−1.232), CCL2 (1.651), andHMOX1 (1.320) were all significantly differentially expressed.Each of these markers plays a distinct role in ECM binding orcytoskeletal remodeling during cell locomotion. These genes,among others, may drive the changes in cell motility followingMoS2_NIR treatment. For example, GREM1 is known to stim-ulate cellular motility and induce potent angiogenic responsein vivo (34, 35). Due to its role in breast cancer motility, invasion,and angiogenesis, GREM1 is considered an attractive moleculartarget in cancer therapeutics. Suppression of GREM1 expressiondue to MoS2_NIR treatment emphasizes its therapeutic poten-tial in cancer. In addition, MoS2_NIR treatment upregulatedHMOX1, which has shown to inhibit migration and cell invasionin breast cancer. It is important to note that spatial rearrange-ment of cells is necessary for angiogenesis. Based on these re-sults, we therefore believe that MoS2 and MoS2_NIR treatmentmay limit the angiogenic potential of cells involved in vasculardevelopment or wound-healing responses by modulating cellularmotility.

MoS2 and Concurrent NIR Exposure Modulate IntegrinSignalingThe reduction in cell motility due to MoS2 and MoS2_NIRtreatment might be affected by integrin signaling (Fig. 5A).Integrin belongs to a glycoprotein family consisting of a non-covalently linked α- and β-subunits. They form heterodimers uponinteractions with ECM components and play critical roles in signal

transduction and functions such as cellular migration, differenti-ation, proliferation, and apoptosis. One of the potential ap-proaches to investigate the interaction between MoS2 nanosheetsand integrin molecules is to quantify the amount of integrin beta-1(CD29) using flow cytometry. Beta-1 integrins recognize the se-quence R–G–D in a wide array of ligands and can form hetero-dimers with integrin alpha-7. Integrin alpha-7/beta-1 is known toregulate cell adhesion, matrix deposition, cell motility, and an-giogenesis. The flow cytometry data showed a significant decreasein unbound integrin beta-1 levels in cells treated with MoS2nanosheets (Fig. 5B and SI Appendix, Fig. S8), thus indicating thatnanosheets inhibit integrin function. Although the flow experi-ments support our primary hypothesis that integrin signaling isinvolved, antibody blocking and knock-down studies are appro-priate methods to further confirm this hypothesis.To identify a potential mechanism by which MoS2 nanosheets

regulate cellular motility, we looked at another important GOterm that showed a high number of differentially expressedgenes: cell adhesion (GO:0007155) (Fig. 5C). A large number ofgenes in this GO term were down-regulated (23 of 28) due toMoS2_NIR treatment compared to control. Some of the prom-inent genes in this GO term are related to extracellular matrixproteins (COL11A1, COL4A1, and FN1), cell–ECM adhesionreceptors (ITGA11, ITGA7, IGFBP2, IGFBP5, NID2, andHAPLN3), and cytoskeletal rearrangement (ACTA2, ACTG2,MYH9, and MYH10) (SI Appendix, Fig. S9). In particular, thedown-regulation of integrin subunits (ITGA7, log2fold: −1.772and ITGA11, log2fold: −1.405) in conjugation with the cyto-skeletal protein (ACTA2, log2fold: −1.506) indicates that MoS2nanosheets may be interfering with surface receptor-mediatedfocal adhesion formation (Fig. 5D).To further validate this hypothesis, we quantified the protein

synthesis of integrin alpha-7 (ITGA7), focal adhesion kinase(FAK), smooth muscle actin (ACTA2), vinculin, and paxillin(Fig. 5E). FAK, vinculin, and paxillin are known to play key rolesin integrin-dependent cellular adhesion, speeding, and motility(36, 37). The results indicate the ITGA7 synthesis was significantlydecreased in cells treated with MoS2 and MoS2_NIR. In-terestingly, no significant difference was observed due to photo-thermal modulation on ITGA7 production. The trend in ITGA7expression in different conditions is similar to results observed inwound-healing experiments, indicating the influence of MoS2 andMoS2_NIR on cellular migration. The expression of other proteinsinvolved in focal adhesion such as FAK, vinculin, paxillin, andACTA2 showed significant decrease in expression in MoS2_NIRcompared to control (hMSCs). To further visualize the effect ofMoS2 nanosheets on focal adhesion and subsequent cellularspreading, the cellular area was measured after treating with MoS2nanosheets (SI Appendix, Fig. S1). We noted a gradual decrease incellular spreading with an increase in MoS2 concentration, in-dicating reduced integrin-mediated cell adhesion.

ConclusionsChemically exfoliated MoS2 is an emerging biomaterial withunique photoresponsive ability. We investigated the biologicalinteractions of MoS2 nanosheets with proteins and cells. Thehigh surface area of MoS2 nanosheets facilitates protein ad-sorption and cellular adhesion/internalization. Once localized tothe cell surface, the MoS2 nanosheets can be exploited with NIRlight to generate thermal stimulation. The effect of MoS2nanosheets and subsequent stimulation with NIR light were in-vestigated via RNA-seq to provide a global and unbiased snap-shot of gene expression of the cells. No significant effect of NIRexposure alone was observed on gene expression of hMSCs.However, treatment with MoS2 nanosheets and subsequent stim-ulation with NIR light influenced ∼103 and 157 genes in hMSCs,respectively. Most of these genes were related to cellular migra-tion and wound healing. Further validation using a wound-healing

13336 | www.pnas.org/cgi/doi/10.1073/pnas.1914345117 Carrow et al.

Dow

nloa

ded

by g

uest

on

Aug

ust 6

, 202

0

Page 9: Photothermal modulation of human stem cells using light-responsive … · Photothermal modulation of human stem cells using light-responsive 2D nanomaterials James K. Carrowa,1 ,

A

APLP1BST1

CCL2

COL12A1

COL16A1

COL1A1

COL5A1

COL5A3

COL8A1

COL8A2

CXCL8

DAG1

FBN1

FN1FZD7

GPNMB

GREM1

HAPLN1HAPLN3

IGFBP2

ITGA11ITGA7

JAM2

JUP

LIMS2

LPXN

MCAM

MYH10 MYH9

NID2

PCDH10

PLXND1

PVR

SAA1

SIRPA

SPP1

TEK

TENM2

THBS1

TINAGL1

TNFSF4TPM1

0

1

2

3

4

5

−2 0 2

-Log

10(p

-adj

ust)

Cell adhesion (GO:0007155)

Log2(hMSCs_MoS2_NIR/hMSCs)

C D

B

ACTA2

0

500

0

500

0

500

0

500

0

500

0

500

0

500

0

500

5 kb

ITGA7

0

60

0

60

0

60

0

60

0

60

0

60

0

60

0

60

5 kb

hMSCs

hMSCs_MoS2

hMSCs_NIR

hMSCs_MoS2_NIR

RPM RPM

ITGA7

GAPDH

MoSNIR

FAK

- + - +- +- +

GAPDH

E

0.0

0.5

1.0

1.5

Nor

mal

ised

Inte

nsity

(ITG

A7/G

APD

H)

***

hMSCs

hMSCs_

NIR

hMSCs_

MoS 2

HMSCs_MoS 2

_NIR

0.0

0.5

1.0

1.5

Nor

mal

ised

Inte

nsity

(FAK

/GAP

DH

)

*

0.0

0.5

1.0

1.5

Nor

mal

ised

Inte

nsity

(AC

TA2/

GAP

DH

)

*

0.0

0.5

1.0

1.5

2.0

Nor

mal

ised

Inte

nsity

(Vin

culin

/GAP

DH

)

*

0.0

0.5

1.0

1.5

Nor

mal

ised

Inte

nsity

(Pax

illin/

GAP

DH

)

*

hMSCs

hMSCs_

NIR

hMSCs_

MoS 2

HMSCs_MoS 2

_NIR

hMSCs

hMSCs_

NIR

hMSCs_

MoS 2

HMSCs_MoS 2

_NIR

hMSCs

hMSCs_

NIR

hMSCs_

MoS 2

HMSCs_MoS 2

_NIR

hMSCs

hMSCs_

NIR

hMSCs_

MoS 2

HMSCs_MoS 2

_NIR

ACTA2

Paxillin

Vinculin

MoSNIR - +- +

- + - +

104 105 1060

30

60

90

120

CD29 Alexa Fluor® 647

Cou

nts

(a.u

)

hMSCs

hMSCs_MoS2

0.0

0.3

0.6

0.9

1.2

1.5

Nor

mal

ized

inte

sity

(a.u

.) **

hMSCs

hMSCs_

MoS 2

MoS2 inhibit integrin binding

ECM

Integrins(ITGA7)

FAKFAK

Actin (ACTA 2)

Paxillin

Vinculin

Actin (ACTA 2)

Fig. 5. Role of integrin expression on cell adhesion due to NIR, MoS2, and MoS2_NIR treatment. (A) Schematic showing the potential interaction betweenMoS2 nanosheets and integrin molecules at the cell surface. (B) Quantification of unbound integrin beta-1 (CD29) in cells treated with and without MoS2nanosheets. (C) A volcano plot for the cell adhesion (GO:0007155), gray: all of the expressed genes, blue: genes associated with the GO term with no sig-nificant change in expression, red: genes associated with the GO term that show significant difference in expression due to treatment. (D) Gene track showingnormalized mRNA expression of integrin alpha-7 (ITGA7) and smooth muscle alpha-2 actin (ACTA2) for hMSCs, hMSCs_NIR, hMSCs_ MoS2, and hMSCsMoS2_NIR. (E) Protein expression level of ITGA7, FAK, vinculin, paxillin, and smooth muscle actin (ACTA2) determined using Western blot for hMSCs,hMSCs_NIR, hMSCs_ MoS2, and hMSCs MoS2_NIR. GAPDH (glyceraldehyde-3-phosphate dehydrogenase) is internal control. *P < 0.05, ***P < 0.001.

Carrow et al. PNAS | June 16, 2020 | vol. 117 | no. 24 | 13337

ENGINEE

RING

Dow

nloa

ded

by g

uest

on

Aug

ust 6

, 202

0

Page 10: Photothermal modulation of human stem cells using light-responsive … · Photothermal modulation of human stem cells using light-responsive 2D nanomaterials James K. Carrowa,1 ,

assay, integrin signaling, and formation of focal adhesion kinaseconfirmed the role of MoS2 and photothermal modulation oncellular functions. Overall, the combination of MoS2 and NIRlight may provide a novel tool to modulate cellular activityfor potential applications in regenerative medicine and cancertherapeutics.

Materials and MethodsSee SI Appendix for detailed materials and methods. Detailed methods fornanoparticle characterization, in vitro studies, and RNA-seq can be found inSI Appendix, Materials and Methods.

Synthesis and Characterization. The bulk MoS2 was chemically exfoliatedfollowing previously reported protocols to obtain MoS2 nanosheets (3, 18,19). The structure of MoS2 nanosheets were evaluated using XRD (Bruker D8Advanced), AFM (Bruker Dimension Icon Nanoscope), TEM (JEOL JEM-2010),XPS (Omicron XPS system with Argus detector), Raman spectroscopy (Lab-Ram HR confocal Raman microscope; Horiba Inc.), DLS (Zetasizer Nano ZS,Malvern Instrument), and zeta potential. Cellular interactions of MoS2nanosheet was determined using hMSCs (obtained from Tissue Culture Core,Institute of Regenerative Medicine, Texas A&M University Health ScienceCenter). The following in vitro studies were performed: metabolic activity(Alamar Blue assay), cell cycle (propidium iodide), apoptosis assay (AnnexinV), and cytosketal staining (actin-phalloidin and nucleus-DAPI staining).

RNA-Seq and Analysis. RNA-seq was performed on hMSCs-treated with NIR(20 mW/cm2), MoS2 nanosheet, and both, using a Nova seq platform (Illu-mina Nova sEq. 6000) utilizing TruSeqRNA preparation and 75 paired-endread length. Sequenced reads were aligned to the human reference genome

(hg38) using a RNA-seq aligner, Spliced Transcripts Alignment to a Reference(STAR) (26). Expression of a gene was determined by counting the number ofuniquely mapped reads overlapping the coding exons normalized by genelength in FPKM. Genes expressed with >1 FPKM in at least half of thesamples of any condition were only used for the analysis. The gene ex-pression read counts were modeled as a negative binomial distribution inGLMs (27) to determine the DEGs. All of the analysis was performed using R.The GO enrichment analysis was done using GOStats Bioconductor package(29). For network formation (Cytoscape) (31) and GO term enrichment(GeneMANIA and ClueGO), only genes with FDR-adjusted P < 0.05 wereselected. REVIGO (32) was performed to visualize GO clustering. Westernanalysis was performed using the iBind system (iBind; Invitrogen).

Statistical Analysis.Determination of statistical significance betweenmultiplegroups was completed via ANOVAwith the Tukey method. Significant P valueswere considered <0.05 unless otherwise noted. All analysis was completed inGraphPad Prism.

Data Availability. The data reported in this paper have been deposited in theGene Expression Omnibus (GEO) database, https://www.ncbi.nlm.nih.gov/geo (accession no. GSE141456).

ACKNOWLEDGMENTS. A.K.G. acknowledges financial support from theNational Institute of Biomedical Imaging and Bioengineering of the NationalInstitutes of Health Director’s New Innovator Award (DP2 EB026265). Wethank Dr. Holly Gibbs for her assistance with experiments utilizing the NIRlaser. The content is solely the responsibility of the authors and does notnecessarily represent the official views of the funding agency. Some of theschematics are prepared using https://biorender.com/.

1. K. S. Novoselov, A. Mishchenko, A. Carvalho, A. H. Castro Neto, 2D materials and van

der Waals heterostructures. Science 353, aac9439 (2016).2. D. Chimene, D. L. Alge, A. K. Gaharwar, Two-dimensional nanomaterials for bio-

medical applications: Emerging trends and future prospects. Adv. Mater. 27,

7261–7284 (2015).3. M. Chhowalla et al., The chemistry of two-dimensional layered transition metal di-

chalcogenide nanosheets. Nat. Chem. 5, 263–275 (2013).4. A. K. Gaharwar et al., 2D nanoclay for biomedical applications: Regenerative medi-

cine, therapeutic delivery, and additive manufacturing. Adv. Mater. 31, e1900332

(2019).5. T. Liu et al., Drug delivery with PEGylated MoS2 nano-sheets for combined photo-

thermal and chemotherapy of cancer. Adv. Mater. 26, 3433–3440 (2014).6. S. S. Chou et al., Chemically exfoliated MoS2 as near-infrared photothermal agents.

Angew. Chem. Int. Ed. Engl. 52, 4160–4164 (2013).7. D. J. Late et al., Sensing behavior of atomically thin-layered MoS2 transistors. ACS

Nano 7, 4879–4891 (2013).8. B. Radisavljevic, A. Radenovic, J. Brivio, V. Giacometti, A. Kis, Single-layer MoS2

transistors. Nat. Nanotechnol. 6, 147–150 (2011).9. K. Jin, L. M. Xie, Y. Tian, D. M. Liu, Au-modified monolayer MoS2 sensor for DNA

detection. J. Phys. Chem. C 120, 11204–11209 (2016).10. E. P. Nguyen et al., Electronic tuning of 2D MoS2 through surface functionalization.

Adv. Mater. 27, 6225–6229 (2015).11. R. F. Frindt, Single crystals of Mos2 several molecular layers thick. J. Appl. Phys. 37,

1928 (1966).12. P. Joensen, R. F. Frindt, S. R. Morrison, Single-layer Mos2.Mater. Res. Bull. 21, 457–461

(1986).13. A. Schumacher, L. Scandella, N. Kruse, R. Prins, Single-layer Mos2 on mica–Studies by

means of scanning force microscopy. Surf. Sci. 289, L595–L598 (1993).14. M. K. Jaiswal, K. A. Singh, G. Lokhande, A. K. Gaharwar, Superhydrophobic states of

2D nanomaterials controlled by atomic defects can modulate cell adhesion. Chem.

Commun. (Camb.) 55, 8772–8775 (2019).15. M. K. Jaiswal et al., Vacancy-driven gelation using defect-rich nanoassemblies of 2D

transition metal dichalcogenides and polymeric binder for biomedical applications.

Adv. Mater. 29, 1702037 (2017).16. W. Z. Teo, E. L. K. Chng, Z. Sofer, M. Pumera, Cytotoxicity of exfoliated transition-

metal dichalcogenides (MoS2, WS2, and WSe2 ) is lower than that of graphene and its

analogues. Chemistry 20, 9627–9632 (2014).17. J. H. Appel et al., Low cytotoxicity and genotoxicity of two-dimensional MoS2 and

WS2. ACS Biomater. Sci. Eng. 2, 361–367 (2016).18. G. Eda et al., Photoluminescence from chemically exfoliated MoS2. Nano Lett. 11,

5111–5116 (2011).

19. L. Yuwen et al., Rapid preparation of single-layer transition metal dichalcogenidenanosheets via ultrasonication enhanced lithium intercalation. Chem. Commun.(Camb.) 52, 529–532 (2016).

20. P. Cheng, K. Sun, Y. H. Hu, Memristive behavior and ideal memristor of 1T phaseMoS2 nanosheets. Nano Lett. 16, 572–576 (2016).

21. O. K. Gasymov, B. J. Glasgow, ANS fluorescence: Potential to augment the identifi-cation of the external binding sites of proteins. Biochim. Biophys. Acta 1774, 403–411(2007).

22. C. C. Fleischer, C. K. Payne, Secondary structure of corona proteins determines the cellsurface receptors used by nanoparticles. J. Phys. Chem. B 118, 14017–14026 (2014).

23. L. Shang, K. Nienhaus, G. U. Nienhaus, Engineered nanoparticles interacting withcells: Size matters. J. Nanobiotechnology 12, 5 (2014).

24. R. Kurapati et al., Enzymatic biodegradability of pristine and functionalized transitionmetal dichalcogenide MoS2 nanosheets. Adv. Funct. Mater. 27, 1605176 (2017).

25. A. E. Nel et al., Understanding biophysicochemical interactions at the nano-bio in-terface. Nat. Mater. 8, 543–557 (2009).

26. A. Dobin et al., STAR: Ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21(2013).

27. M. I. Love, W. Huber, S. Anders, Moderated estimation of fold change and dispersionfor RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

28. J. K. Carrow et al., Widespread changes in transcriptome profile of human mesen-chymal stem cells induced by two-dimensional nanosilicates. Proc. Natl. Acad. Sci.U.S.A. 115, E3905–E3913 (2018).

29. S. Falcon, R. Gentleman, Using GOstats to test gene lists for GO term association.Bioinformatics 23, 257–258 (2007).

30. P. Shannon et al., Cytoscape: A software environment for integrated models of bio-molecular interaction networks. Genome Res. 13, 2498–2504 (2003).

31. M. Kohl, S. Wiese, B. Warscheid, Cytoscape: Software for visualization and analysis ofbiological networks. Methods Mol. Biol. 696, 291–303 (2011).

32. F. Supek, M. Bošnjak, N. Skunca, T. Smuc, REVIGO summarizes and visualizes long listsof gene ontology terms. PLoS One 6, e21800 (2011).

33. D. W. Howell, C. W. Peak, K. J. Bayless, A. K. Gaharwar, 2D nanosilicates loaded withproangiogenic factors stimulate endothelial sprouting. Adv. Biosyst. 2, 1800092 (2018).

34. S. Mitola et al., Gremlin is a novel agonist of the major proangiogenic receptorVEGFR2. Blood 116, 3677–3680 (2010).

35. G. S. Karagiannis, A. Berk, A. Dimitromanolakis, E. P. Diamandis, Enrichment mapprofiling of the cancer invasion front suggests regulation of colorectal cancer pro-gression by the bone morphogenetic protein antagonist, gremlin-1. Mol. Oncol. 7,826–839 (2013).

36. C. Wu, Focal adhesion: A focal point in current cell biology and molecular medicine.Cell Adhes. Migr. 1, 13–18 (2007).

37. X. Zhao, J.-L. Guan, Focal adhesion kinase and its signaling pathways in cell migrationand angiogenesis. Adv. Drug Deliv. Rev. 63, 610–615 (2011).

13338 | www.pnas.org/cgi/doi/10.1073/pnas.1914345117 Carrow et al.

Dow

nloa

ded

by g

uest

on

Aug

ust 6

, 202

0